135
הטכניון לישראל טכנולוגי מכוןTechnion – Israel Institute of Technology הטכניון ספריותThe Technion Libraries בית הספר ללימודי מוסמכים ע" ש ארווין וג' ואן ג' ייקובסIrwin and Joan Jacobs Graduate School © All rights reserved This work, in whole or in part, may not be copied (in any media), printed, translated, stored in a retrieval system, transmitted via the internet or other electronic means, except for "fair use" of brief quotations for academic instruction, criticism, or research purposes only. Commercial use of this material is completely prohibited. © כל הזכויות שמורות להעתיק אין) כלשהי במדיה( , להדפיס, לתרגם, מידע במאגר לאחסן, באינטרנט להפיץ, או זה חיבור ממנו חלק כל, למעט" הוגן שימוש" לימוד למטרות החיבור מן קצרים בקטעים, הוראה, ביק ו ר או ת מחקר. בהחלט אסור זה בחיבור הכלול בחומר מסחרי שימוש. © Technion - Israel Institute of Technology, Elyachar Central Library

Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

מכון טכנולוגי לישראל –הטכניון

Technion – Israel Institute of Technology

ספריות הטכניוןThe Technion Libraries

ייקובס'ג ואן'וג ארווין ש"ע מוסמכים ללימודי הספר בית

Irwin and Joan Jacobs Graduate School

© All rights reserved

This work, in whole or in part, may not be copied (in any media), printed,

translated, stored in a retrieval system, transmitted via the internet or other electronic means, except for "fair use" of brief quotations for academic

instruction, criticism, or research purposes only. Commercial use of this material is completely prohibited.

© שמורות הזכויות כל

חיבור זה או , להפיץ באינטרנט, לאחסן במאגר מידע, לתרגם, להדפיס, )במדיה כלשהי(אין להעתיק

ת או רוביק, הוראה, בקטעים קצרים מן החיבור למטרות לימוד "שימוש הוגן"למעט , כל חלק ממנו .שימוש מסחרי בחומר הכלול בחיבור זה אסור בהחלט. מחקר

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 2: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

Elucidation of aminoglycosides modes of activity

in eukaryotes: towards improved therapeutic

derivatives

Moran Shalev Ben-Ami

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 3: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

Elucidation of aminoglycosides modes of activity

in eukaryotes: towards improved therapeutic

derivatives

Research Thesis

In Partial Fulfillment of the Requirements

For the Degree of

Doctor of Philosophy

Moran Shalev Ben-Ami

Submitted to the senate of the

Technion - Israel Institute of Technology

Tammuz 5772 Haifa June 2013

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 4: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

This research was conducted under the supervision of

Prof. Timor Baasov and Prof. Noam Adir

At the Schulich Faculty of Chemistry, Technion, Israel

The Generous Financial Help Of the Technion and Mr. Seymour Schulich Is

Gratefully Acknowledged

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 5: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

List of Publications

Publication in scientific journals

Shalev M., Kondo J., Kopelyanskiy D., Jaffe C.L., Adir N., and Baasov T. (2013).

Identification of the molecular attributes required for Aminoglycoside activity

against Leishmania. Proceedings of the National Academy of Sciences USA.

Accepted. IF 9.681; Ranked 3 out of 56 in Multidisciplinary Sciences.

Hariton A., Shalev M., Adir N., Belausov E. and Altstein M. (2013). Structural and

functional differences between cloned and expressed Heliothis peltigera and

Spodoptera littoralis PK/PBAN receptors. Biochimica et Biophysica Acta – general

subjects. Accepted. doi: 10.1016/j.bbagen.2013.06.041. IF 5.000; Ranked 89 out of

290 in Biochemistry & Molecular Biology. Performed the structural studies

including homology model building and structural analysis.

Xue X., Mutyam V., Tang L.P., Biswas S., Du M., Jackson L.A., Dai Y., Belakhov

V., Shalev M., Chen F., Schacht J., Bridges R., Baasov T., Hong J., Bedwell D.M.,

and Rowe S.M. (2013). Synthetic Aminoglycosides Efficiently Suppress CFTR

Nonsense Mutations and Are Further Enhanced by the CFTR Potentiator

Ivacaftor. American Journal of Respiratory Cell and Molecular Biology. Under

review. IF 5.125; Ranked 76 out of 290 in Biochemistry & Molecular Biology.

Ranked 7 out of 50 in Respiratory System. Performed AG monitoring in mice

serum samples.

Shalev M., Kandasamy J., Skalka N., Belakhov V., Rosin-Arbesfeld R., and Baasov

T. (2012). Development of generic immunoassay for the detection of a series of

aminoglycosides with 6′-OH group for the treatment of genetic diseases in

biological samples. Journal of Pharmaceutical and Biomedical Analysis. 75:33–40.

IF 2.979; Ranked 20 out of 73 in Analytical Chemistry.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 6: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

Presentations in scientific conferences

Oral and Poster presentations: Shalev M., Kondo J., Kopelyanskiy D., Jaffe C.L.,

Adir N., and Baasov T. Identification of the molecular attributes required for

Aminoglycoside activity against Leishmania. Eurocarb 17, Tel-Aviv, 2013.

Oral and Poster presentations: Shalev M., Kondo J., Kopelyanskiy D., Jaffe C.L.,

Adir N., and Baasov T. Structural insights of aminoglycoside activity in protozoan

parasites. The Israeli Medicinal Chemistry Conference, Rehovot, June 2013.

Oral presentation: Shalev M., Kondo J., Kopelyanskiy D., Jaffe C.L., Adir N., and

Baasov T. Identification of the molecular attributes required for Aminoglycoside

activity against Leishmania. ICA (Israel Crystallography Association), Haifa, May

2013.

Poster presentation: Shalev M., Kondo J., Kopelyanskiy D., Jaffe C.L., Adir N., and

Baasov T. Structural insights of aminoglycoside activity in protozoan parasites.

(Awarded 1st prize) ICS (Israel Chemical Society) meeting, Tel Aviv, Feb 2013.

Poster presentation: Shalev M., Adir N., and Baasov T. Structural insights into

the mechanism of aminoglycoside dependent stop-codon read-through in human

ribosomes. ICS (Israel Chemical Society) meeting, Tel-Aviv, Feb 2011.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 7: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

Table of Contents

List of Figures and Tables.............................................................................................

List of Publications……………………………………………………………………

Abstract ............................................................................................................................... 1

List of Abbreviations .......................................................................................................... 3

1. Introduction ..................................................................................................................... 6

1.1 Aminoglycosides....................................................................................................... 6

1.2 Aminoglycosides Mechanism of Action in Bacteria ................................................ 7

1.3. Aminoglycosides and Genetic Diseases ................................................................ 10

1.4. The Read-Through Mechanism ............................................................................. 12

1.5 Aminoglycosides Activity against Parasitic Protozoa - Leishmania ...................... 16

1.6 Aminoglycosides Toxicity ...................................................................................... 17

1.7 Challenges in the Design of Novel Drugs Targeting the Eukaryotic Ribosome .... 18

1.7.1 Development of Read-Through Inducers ......................................................... 18

1.7.2 Development of Anti-Protozoal Derivatives ................................................... 21

1.8 Aminoglycosides and the Eukaryotic Ribosome - Current Knowledge and

Alternative Model Systems ..................................................................................... 22

2. Research Goals and Perspectives .................................................................................. 27

3. Materials and Methods ................................................................................................. 29

3.1 Materials ................................................................................................................. 29

3.2. General Techniques ............................................................................................... 29

3.2.1 X-ray Analysis ................................................................................................. 29

3.2.2 NMR, MS and TLC ......................................................................................... 29

3.3 RNA Purification and Crystallization ..................................................................... 30

3.3.1 RNA Purification ............................................................................................. 30

3.3.2 RNA Annealing ............................................................................................... 31

3.3.3 Crystallization .................................................................................................. 31

3.4. RNA-models: Crystal Handling, Data Collection, Structure Determination and

Refinement .................................................................................................................... 32

3.4.1 Standard Crystallization Procedures and Data Handling ................................. 32

3.4.2 Additional Crystallization Techniques (Seeding, Soaking) ............................. 32

3.5 Yeast 80S Ribosomes Crystallization and Crystal Structure Determination .......... 33

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 8: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

3.6 Leishmania Cell Culture and Promastigote Viability Assay (IC50). ....................... 35

3.7 The Development of a Highly Sensitive Immunoassay for 6'-OH AG Detection .. 36

3.7.1 Preparation of Polyclonal Abs ......................................................................... 36

3.7.1.1 Preparation of 6'-(R)-methyl-2',3-diazido-1-N-[5-oxopentanoic acid]- ... 37

paromamine (compound B, Figure 3.1). ............................................................... 37

3.7.1.2 Preparation of Immunization Conjugate (compound E, Figure 3.1). ....... 37

3.7.1.3 Immunization and Antiserum Production. ................................................ 38

3.7.2 The Development of a Highly Sensitive Immunoassay ................................... 38

3.7.2.1 Preparation of NB82-OVA Coating Antigen. ........................................... 38

3.7.2.2 Preparation of NB82/84/124-GTA-OVA Coating Antigens .................... 39

3.7.2.3 NB82 Indirect Competitive ELISA .......................................................... 40

3.7.2.4 Cross Reactivity Experiments ................................................................... 40

3.7.2.5 NB82/84/124 Indirect Competitive ELISA .............................................. 41

3.7.2.6 Data Analysis ............................................................................................ 41

3.7.3 Determination of NB Compounds Levels in Mice Serum ............................... 42

4. Results and Discussion ................................................................................................. 43

4.1 Deciphering Aminoglycosides Mechanisms of Action in Eukaryotes ................... 43

4.1.1 rRNA Purification, Complex Generation and Crystallization ......................... 44

4.1.2 Crystal Structure Determination of Human A-site rRNA Complexes with

AG ............................................................................................................................. 46

4.1.2.1 The Double A-site Model ......................................................................... 46

4.1.2.2 The Single A-site Model ........................................................................... 62

4.1.3 Crystal Structure Determination of 80S Yeast Ribosomes in Complex with

AG ............................................................................................................................. 66

4.1.4 Leishmania A-site Crystal Structure Determination ........................................ 69

4.1.4.1 The Binding Mode of G418 to the Leishmanial A-site - ‘ON’ State........ 75

4.1.4.2. The Binding Mode of Apramycin to the Leishmanial A-site - ‘OFF’

State....................................................................................................................... 77

4.1.5. In-vitro Inhibition of L. donovani and L. major Promastigote Growth by

AG. ............................................................................................................................ 79

4.1.6. Aminoglycosides Mechanisms of Action in Leishmania ............................... 82

4.2. The Development and Employment of a Highly Sensitive Immunoassay for the

Detection of AG in Biological Samples................................................................. 84

4.2.1 Preparation of Immunization Conjugate .......................................................... 86

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 9: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

4.2.2 Development of an Indirect Competitive ELISA ............................................ 88

4.2.3 Development of Heterologous-Assays ............................................................ 93

4.2.4 Determination of Selected NB Compounds Serum Levels in Mice ................ 94

5. Summary and Conclusions ........................................................................................... 98

Appendix ........................................................................................................................... 99

References ....................................................................................................................... 103

I ............................ ...........................................................................................תקציר מחקר

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 10: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

List of Figures and Tables

Figures:

Figure 1.1: Chemical structures of natural AG derivatives……………………………..…… 7

Figure 1.2: Molecular glance at the bacterial decoding site…………………………...…..… 9

Figure 1.3: The molecular basis of aminoglycosides action in the bacterial ribosome…….... 9

Figure 1.4: The read-through mechanism…………………………………………......…… 11

Figure 1.5: Schematic view of translation and translation termination…………..………... 14

Figure 1.6: A molecular glance at the decoding site conformation upon RF binding to the

bacterial A-site………………………………………………………………...……. 15

Figure 1.7: Bacterial vs. Human mithochondrial A-site……………………………..….… 17

Figure 1.8: Structures of novel semi-synthetic AG that induce translational read-through of

premature stop codons………………………………………………………...……. 21

Figure 1.9: Structures of novel semi-synthetic AG exhibiting some anti-protozoal

properties……………………………………………………………………...……. 22

Figure 1.10: Two dimensional representation of the bacterial vs. eukaryotic cytoplasmic

decoding sites (A-sites)……………………………………………………….…….. 25

Figure 1.11: Superposition of Paromomycin bound to a minimal A-site model and to the

70S ribosome apparatus….……………………………………………………….… 26

Figure 1.12: The eukaryotic A-site models used for the structural exploration of AG

binding site……………………………………………………………………….…. 26

Figure 3.1: Synthetic scheme for the preparation of immunization\coating conjugates…… 36

Figure 4.1: The rRNA constructs used to explore AG interactions…………..…………… 43

Figure 4.2: RNA deprotection and purification………………………………………….… 44

Figure 4.3: Crystals emerged from an initial screen of eukaryotic rRNA duplex in complex

with NB54…………………………………………………………………………... 45

Figure 4.4: Crystals obtained from an optimization screen of eukaryotic rRNA duplex in

complex with NB54………………………………………………………………… 46

Figure 4.5: Structures of rRNA crystals that were crystallized in the presence of a potent

read-through inducer – NB54………………………………………………………. 48

Figure 4.6: NB74-human cytoplasmic A-site complex………………………………..…... 53

Figure 4.7: B-factor analysis of NB54-Cyt1 and NB74-Cyt2 …………………………….. 53

Figure 4.8: Crystal packing……………………………………………………………..….. 54

Figure 4.9: A. Crystals Apramycin-eukaryotic-rRNA duplex…………………………..…. 55

Figure 4.10: Crystal packing of the Apra-cyt complex………………………………...….. 56

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 11: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

Figure 4.11: G418-human cytoplasmic A-site complex at pH 5.0…………………………. 58

Figure 4.12: Analysis of the pH 5.0 ‘ON’ state structure………………………………..… 61

Figure 4.13: Crystals emerged from single eukaryotic A-site rRNA duplex in complex

with AG……………………………………………………………………………. 63

Figure 4.14: Apramycin bound to the extended single A-site model…………………….... 63

Figure 4.15: Asymmetric unit and crystal packing of crystals emerged of complexes of

the read-through inducers………………………………………………………..…. 65

Figure 4.16: Crystallization of yeast 80S ribosomes…………………………………….… 67

Figure 4.17: Secondary structures of the bacterial, leishmanial and human cytoplasmic

A-sites………………………………………………………………………………. 69

Figure 4.18: Crystals emerged from crystallization experiments of complexes of the

leishmanial A-site model with G418 and Apramycin…………………………..…. 71

Figure 4.19: 2D and 3D representations of the double A-site complexes as obtained in the

X-ray crystal structures…………………………………………………………..…. 71

Figure 4.20: Electron density maps of the Apra-Leish and G418-Leish structures……….. 72

Figure 4.21: Surface representation of apramycin bound to the double A-site rRNA

construct corresponding to its putative binding site in leishmanial ribosomes…….. 72

Figure 4.22: Crystal structure visualization of G418 or Apramycin - leishmanial A-site

rRNA complexes……………………………………………………………………. 74

Figure 4.23: Description of the contacts between G418 and the leishmanial A-site…….…. 76

Figure 4.24: Description of the contacts between Apramycin and the leishmanial A-site..... 78

Figure 4.25: Surface representations of AG binding sites in leishmanial ribosomes…...…. 82

Figure 4.26: General scheme of AG activities in Leishmania………………………….… 83

Figure 4.27: Chemical structures of standard and semi-synthetic AG derivatives used in

the study…………………………………………………………………………..… 86

Figure 4.28: Antiserum activity…………………………………………………………….. 88

Figure 4.29: Antigen recognition (Chessboard) assay……………………………………… 89

Figure 4.30: Standard curve of NB82 (Homologous assay)………………………………... 89

Figure 4.31: Homologous assay cross reactivity…………………………………………… 92

Figure 4.32: Serum levels of NB84 and NB124 following a single dose IP injection in

male mice……………………………………………………………………...……. 96

Tables:

Table 4.1: Crystal data, data collection and data refinement statistics – NB54…………….. 47

Table 4.2: Crystal data, data collection and data refinement statistics……………………... 50

Table 4.3: Crystal data, data collection and data refinement statistics……………………... 52

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 12: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

Table 4.4: Crystal data, data collection and data refinement statistics for the Apra-cyt

crystal……………………………………………………………………………… 56

Table 4.5: Crystal data, data collection and data refinement statistics ……………………. 59

Table 4.6: Crystal data, data collection and data refinement statistics…………………..… 64

Table 4.7: Crystal data, data collection and data refinement statistics…………………….. 68

Table 4.8: Crystal data, data collection and data refinement statistics…………………….. 73

Table 4.9: In-vitro inhibition of Leishmania promastigotes by natural AG……………… 80

Table 4.10: In-vitro inhibition of Leishmania promastigotes by synthetic AG and

toxicity measurements in HEK293 cells (LC50)………………………………….... 81

Table 4.11: Concentrations at 50% inhibition (I50) and limits of detection (I20) of various

AG using the homologous ELISA with NB82-OVA as a coating antigen....….…. 90

Table 4.12: Concentrations at 50% inhibition (I50) and limits of detection (I20) of

representative members using the heterologous ELISAs…………………………. 94

Table 4.13: Concentrations at 50% inhibition (I50) and limits of detection (I20) of

representative members in mice serum…………………………………………… 95

Table 4.14: Recovery of NB82, NB84 and NB124 from whole blood samples…………... 95

Table I: Crystal screen and initial crystallization conditions determination……………… 99

Table II: Crystal optimization matrix……………………………………………………. 101

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 13: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

1

Abstract

Aminoglycosides (AG) are mostly known as highly potent, broad-spectrum

antibiotics that exert their antibacterial activities by selectively targeting the decoding A-

site of bacterial ribosomes, leading to aberrant protein synthesis. Recent studies

suggested that the action of AG is not limited to bacterial ribosomes only; such studies

demonstrated the ability of some AG to interfere with several aspects of eukaryotic

translation. As such, AG were marked as excellent candidates for the treatment of

nonsense mutations mediated human genetic disorders and as alternative treatments for

infections caused by human parasitic protozoa. Nevertheless, despite the great potential

of using AG for such additional therapeutic purposes, their use is quite restricted due to

relatively high toxicity values in human and the information regarding their mechanisms

of action/toxicity in eukaryotes is rather obscure.

Over the last few years several synthetic derivatives where suggested to overcome

some of the enhanced toxicity issues as well as exhibited some improved activity and

selectivity in eukaryotes; many of these derivatives were synthesized in our lab. These

derivatives bear a great potential to serve as therapeutic candidates. However, the newly

synthesized derivatives also demonstrated the necessity to further understand the

molecular mechanisms in which AG confer their biological activity in eukaryotic cells for

further rational drug design.

The present work aimed at exploring AG mechanism(s) of action in eukaryotes at

the molecular level, by solving the X-ray crystal structures of AG bound to their putative

binding sites in eukaryotes - the eukaryotic ribosome A-site where tRNA selection occurs

at the ribosome. These studies were based on the high conservation of the decoding site

among all living kingdoms and the already published literature regarding their ability to

interfere with eukaryotic translation. We present here the crystal structures of different

AG molecules bound to RNA molecules serving as models of the human ribosomal A-

site at resolutions ranging between 2.5-3.0 Å. In addition, crystal structures of two

different AG molecules bound to a model of the Leishmania ribosomal A-site are also

presented: Geneticin (G418), a potent AG for the treatment of leishmaniasis at a 2.65Å

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 14: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

2

resolution, and Apramycin, shown to be a strong binder to the leishmanial ribosome

lacking an anti-leishmanial activity at 1.4Å resolution. The structural data, coupled with

in-vitro inhibition measurements on two strains of Leishmania, provides for the first time

insight as to the source of the difference in inhibitory activity of different AG. The

combined structural and biological data sets the ground for rational design of new and

more specific AG derivatives as potential therapeutic agents against leishmaniasis and

nonsense mediated genetic disorders.

In addition to the structural studies, a highly sensitive immunoassay for the

determination and quantification of synthetic AG in biological samples was developed.

The assay has been developed by the generation of highly specific polyclonal antibodies in

rabbits. The assay's efficiency and reproducibility have been demonstrated in treated mice

serum samples. Such assay could be of a great use in the further development of these

therapeutic compounds, allowing the monitoring of AG activities in-vivo. Such tools could

also be exploited for testing some additional mechanistic aspects of AG activities in

eukaryotes such as their permeability characteristics and could be of a great help towards

improving their therapeutic indexes.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 15: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

3

List of Abbreviations

2-DOS - 2-deoxystreptamine

2'-ACE - Acid-labile orthoester protecting group

aa - Amino acid

aa-tRNA - aminoacylated tRNA

Ab - Antibody

AG - Aminoglycoside

AHB - (s)-4-amino-2-hydroxybutanoyl

Apra-Leish - Apramycin bound to leishmanial A-site

A-site - aminoacyl tRNA biding site

AT - Ataxia telangiectasia

CB - Carbonate buffer

CD - Circular dichroism

CE - Capillary electrophoresis

CF - Cystic fibrosis

CFTR - Cystic fibrosis transmembrane conductance regulator

Co-Hex - Cobalt Hexammine

DCC - N'-N'-dicyclohexylcarbodiimide

DDW - Double distilled water

DMD - Duchenne muscular dystrophy

DMF - Dimethylformamide

DMSO - Dimethyl sulfoxide

EF - Elongation factor

ELISA - Enzyme-Linked Immunosorbent Assay

ESI - Electrospray ionization

E-site - Exit tRNA binding site

ESRF - European Synchrotron Radiation Facility

FBS - Fetal bovine serum

FIA - Flouroimmunoassays

FRET - Fluorescence resonance energy transfer

G418 - Geneticin

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 16: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

4

G418-Leish - Geneticin bound to leishmanial A-site

G418-Bact - Geneticin bound to bacterial A-site

GAG - Glycosaminoglycan

GC - Gas chromatography

GTA - Glutaraldehyde

H34 - helix 34

H44 - helix 44

H69 - Helix 69

HPLC - High-performance liquid chromatography

HRP - Horseradish Peroxidase

HS - Hürler syndrome

I50 - Sensitivity level (50% Antibody inhibition)

I20 - Limit of detection (20% Antibody inhibition)

IC50 - 50% Growth inhibition values

IP - Intraperitoneal

ITC - Isothermal titration calorimetry

KLH - Keyhole Limpet Hemocyanin

LC50 - 50% Cell growth inhibition values

LD50 - lethal toxicity values

MPD - 2,4-methyl-pentadiol

MPS I-H - Mucopolysaccharidosis type-I-Hurler

MS - Mass spectra

NHS - N-hydroxysuccinimide

NMR - Nuclear magnetic resonance

Os-Hex - Osmiun Hexammine

OVA - Ovalbumin

PAGE - Polyacrylamide gel electrophoresis

PBS - Phosphate buffered saline

PBST - PBS containing Tween-20

PDB - Protein Data Bank

PDR - Physicians Desk Reference

PEG - Polyethylene glycol

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 17: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

5

PMe3 - Trimethylphosphine

PSI - Paul Scherrer Institue

P-site - peptidyl tRNA binding site

PTC - premature termination codon

RF - Release factors

RIA - Radioimmunoassays

ROS - Reactive oxygen species

RRE - Rev responsive element

S.E.M - Standard error mean

SLS - Swiss Light Source

SMA - Spinal muscular atrophy

SMN - Survival motor neuron 1

TAR - Tat responsive element

THF - Tetrahydrofuran

TLC - Thin layer chromatography

TMB - 3,3',5,5'-tetramethyl benzidine

USH - Usher syndrome

VPA - Valoparic acid

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 18: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

6

1. Introduction

1.1 Aminoglycosides

Aminoglycosides (AG) were first established as antibiotics in the 1940s with the

discovery of Streptomycin and are still widely used worldwide for the treatment of

various infectious diseases 1, 2

. Chemically AG are cationic oligosaccharides composed of

between two and five amino sugar rings. At physiological pH, the amino groups are

nearly all protonated, giving the AG a net positive charge 3, 4

. AG can be categorized

structurally into three groups based upon the identity of conserved aminocyclitol-2-

deoxystreptamine (2-DOS) ring around which they are built. The amino sugar moieties

are distributed about the 2-DOS ring in three major substitution patterns: 4-

monosubstituted - Apramycin; 4,5-disubtituted - Neomycin and Paromomycin; and 4,6-

disubsituted - Gentamicins and Kanamycins (Figure 1.1).

The bacterial ribosome is thought to be the primary target of AG containing a 2-

DOS ring, in which they selectively bind to some highly conserved rRNA residues

composing the ribosomal aminoacyl-tRNA binding site (A-site) where tRNA selection

occurs. AG binding modifies the ribosome's ability to effectively select tRNAs; therefore

severely enhance the abundance of miscoding events, resulting in the accumulation of

mistranslated proteins and eventually leading to cell death. In addition to bacterial

ribosomes, AG have been shown to bind numerous RNA constructs such as ribozymes 5,

introns 6, HIV-1 Tat-responsive element (TAR)

7 and HIV-1 Rev-responsive element

(RRE) 8 and modulate their structure and function. Recent studies, demonstrating the

potential of some AG to influence translational processes of eukaryotes also highlighted

these compounds as potential therapeutic candidates for the treatment of a wide variety of

parasite related infections 9 and some genetic disorders caused by nonsense mutations

10-

12. However, despite these promising clinical results, the mechanistic and structural

information regarding AG action in eukaryotes is currently very limited.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 19: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

7

OH OH

NH2 H

OH

OH

R1 R2 R3

Kanamycin A

Tobramycin

NH2 OH

NH2 OH

R1 R2

NeamineRibostamycin

O

OH

NH2

AHB =

IV

OHHO

NH2NHR4

O

O

O

NH2

R3R2

R1

OHO

H2NOH

OH

II

III

I III

R3

H

H

AHBOH

OH OH OH

R4

H

H

AHB

O

ONH2

OH

OH

H2N

OHO

O

OH

NH2

NHR3

OH

O

O

R1

HOR2

H2N

III

NH2 H H AHBHNH2 H H

H

HNH2

OH

OH

OH

Paromomycin

Neamine

Ribostamycin

Neomycin B

6' 6'

5

4

5

Butirosin A

ButirosinAmikacin

4

ArbekacinDibekacin

Neomycin B

Paromomycin

HNH2 OH OHKanamycin B

NH2

NHCH3 CH3

NH2 CH3

H

R1 R2

Gentamicin C1

Gentamicin C2

Gentamicin C1A

I

HO

NH2

NH2

O

O

OR1

H2N

OHO

HN OH

R2

II

III

6'

4

6

R3

R4 R3 R4

H H

HH

H H

Geneticin (G418) OH CH3 OH OH

I

HO

NH2

NH2

OH

O

O

H2N

IIOO

O

OH

H2NHO

OHHN

CH3

OH

4Apramycin

Figure 1.1: Chemical structures of natural AG derivatives. The 2-DOS conserved ring is highlighted in

blue.

1.2 Aminoglycosides Mechanism of Action in Bacteria

The antibacterial mechanism of action of AG has been well characterized and it

was discovered in the late 1980s that their molecular target is the 16S rRNA which is

located in the small ribosomal subunit (30S) 13

. The ribosome is a large macromolecular

enzyme that catalyzes the sequential addition of amino acids to a growing polypeptide

chain, using mRNA as a template and aminoacylated tRNAs (aa-tRNA) as substrates. In

all organisms, the ribosome consists of a large and a small subunit, which are referred

according to their rate of sedimentation. During the subsequent process of translation,

both subunits act in concert, which involves the movement of mRNA and all three tRNAs

by precisely one codon with respect to the ribosome. The entire process also depends on

several extrinsic protein factors and the hydrolysis of GTP. The ribosome catalytic site

consists of three tRNA-binding sites, designated A (aminoacyl), P (peptidyl) and E (exit),

after their respective tRNA substrates. Recent structural evidence indicate that these three

binding sites are all composed of RNA elements from at least two different ribosomal

domains (Figure 1.2).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 20: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

8

During decoding, a critical step in aminoacyl-tRNA selection is based on the

formation of a mini-helix between the codon of the mRNA and the anti-codon of the

cognate aa-tRNA. This step is believed to occur within the ribosomal A-site, located in

the small ribosomal subunit. Data from chemical protection 14

, cross linking 15

, and

genetic analysis 16

suggest that the decoding site is a region around the ribosomal A-site

that includes helix 44 (H44), the 530 loop, and helix 34 (H34) of 16S RNA. The correct

recognition of each A-site codon by the anticodon of the corresponding aa-tRNA requires

delicate discrimination against non-cognate and near-cognate tRNAs. The high level of

accuracy of translation by the ribosome is thought to be a combination of the initial

codon-anticodon interactions and a very sophisticated proof-reading ability.

Following the determination of the bacterial ribosome structure by X-ray

crystallography, Ramakrishnan and co-workers 17

suggested an atomic level mechanism

of tRNA recognition by the ribosome. In general, the binding of mRNA and cognate

tRNA in the A-site induces two highly conserved bases, A1492 and A1493, to flip out

from the internal loop of H44 (Figure 1.3A). In their new conformations, these residues

tightly interact with the first two bases of the codon-anticodon helix and are therefore

able to monitor and discriminate between Watson-Crick base pairing and mismatches. In

the absence of cognate codon-anticodon mini-helix the two conserved adenines can

occupy one of 2 possible conformations in which they could either both be folded back

within H44 (Figure 1.3B) or only one residue is flipped out (A1493) where the other

(A1492) is directed inside the helical core held by stacking interactions with an adjacent

adenine residue located at the large ribosomal subunit (Figure 1.3C) 18

. When one of

these two conformations occurs the ribosome occupies a rather open conformation and

the translation process is hindered. These possible conformational states were later been

denoted by Westhof and co-workers 19

as 'OFF' (hindered) and 'ON' states and could be

seen as a molecular switch that irreversibly decides whether translation will continue.

The binding of AG to the bacterial A-site seem to stabilize the 'ON' conformation even in

the absence of cognate tRNA-mRNA complex (Figure 1.3D). Thus, the affinity of the A-

site for a near-cognate mRNA-tRNA complex is increased upon AG binding, preventing

the ribosome from efficiently discriminating between near-cognate and cognate

complexes. These interactions reduce the fidelity of normal translational processes

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 21: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

9

leading to the accumulation of mistranslated non-functional proteins in bacterial cells,

eventually leading to cell death.

Figure 1.2: Molecular glance at the bacterial decoding site. The 30S ribosomal subunit is showed at the left

side with three tRNA molecules bound (A-site – green; P-site – Yellow; E-site – Blue) and mRNA

highlighted in black. The A-site tRNA is bound to an EF (elongation factor – light pink) which is an

intrinsic protein participating in the translation elongation process. The actual decoding site is highlighted

in red, and is enlarged in the right side of the figure. The mRNA in the enlarged view is highlighted in

yellow; A-site bound tRNA in green; and the two conserved adenine residues, A1492 and A1493, are

highlighted in blue and red, respectively. The PDB entry for the presented structure is 2WRQ.

Figure 1.3: The molecular basis of aminoglycosides

action in the bacterial ribosome, as visualized from

recent X-ray crystal structures of T.thermophilus and

E.coli. The relevant part of H44 is presented in

cartoon representation (grey); A1492 (blue), A1493

(red), and the conserved A1408 (orange) are

represented in filled ring ball and stick

representation. The various conformational

corresponding to A. 'ON' state conformation, where

A1492 and A1493 are flipped out from the helical

core (PDB ID: 1IBM); B. 'OFF' state conformation,

where both adenines are stacked within the interior

of the rRNA helix (PDB ID: 2B9O); and C.

Alternative 'OFF' state conformation where A1492 is

directed towards the helical core and A1493 is fully

bulged out (PDB ID: 3I9B). D. The 30S particle in

complex with Paromomycin. The unique H-bond

pattern is indicated in blue dashed lines. The A1492

and A1493 bases are flipped out, thus indicating the

'ON' state conformation. Paromomycin is

represented in yellow ball and stick representation

(PDB ID: 1IBL).

A. B.

C. D.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 22: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

10

1.3. Aminoglycosides and Genetic Diseases

AG antibacterial activities as miscoding agents have been extensively investigated

over the last 7 decades. However, nearly 2 decades after the discovery of streptomycin, it

was discovered that AG are also able to suppress premature termination codons (PTCs),

restoring full-length protein production in Escherichia coli (E. coli) 20

. PTCs are genetic

code mutations usually occurring due to base pair insertions, deletions, or substitutions

that result in the replacement of an amino-acid codon by one of the three universal stop

codons (UAA, UGA or UAG). These mutations generally lead to the production of

truncated, non-functional proteins (Figure 1.4). In humans, PTCs have been linked to

nearly 2,000 genetic disorders, such as Cystic fibrosis (CF), Duchenne muscular

dystrophy (DMD), ataxia-telangiectasia (AT), Hurler syndrome (HS), Rett syndrome,

Usher syndrome (USH), Hemophilia A, Hemophilia B and Tay-Sachs 21, 22

. For many of

these diseases there is presently no effective treatment.

In the last several years, a new therapeutic strategy has suggested the use of PTCs

suppression in the treatment of several PTC induced genetic disorders 23

. Recent

scientific evidence have demonstrated the ability of some natural AG, such as

Gentamicin, G418 and Paromomycin to induce PTC read-through in various eukaryotic

systems 24-26

, via the selective insertion of a near cognate tRNA at the PTC position;

restoring the production of full length functional proteins (Figure 1.4C). These proof-of-

principle studies have suggested the use of such AG as a possible treatment for human

genetic diseases cause by PTCs, and indeed, recent clinical studies performed in Cystic

fibrosis patients carrying one of the CFTR stop mutations indicated Gentamicin's ability

to improve patients' symptoms 27, 28

.

These encoureging results were further exploited for the establishment of several

clinical trials in DMD patients 29

altogether with massive experiments performed in

various in-vitro, ex-vivo and in-vivo systems on DMD 30, 31

, CF 32

, Rett syndrome 33, 34

,

HS 35

, nephrogenic diabetes insipidus 36

, nephropathic cystinosis 37

, retinitis pigmentosa

38, ataxia-telegiectasia

39, spinal muscular atrophy (SMA)

40 and several genetically

induced cancer types 41-43

. The resulting data supported the previous findings and

highlighted AG as possible candidates for PTC therapy. However, it also emphasized the

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 23: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

11

complexity of the mechanisms in which these agents induce their therapeutic effects. For

example, it has been noticed that the sequence context of the PTC affects the efficacy of

mutation suppression (UGA > UAG > UAA). In addition, the nucleotides located

immediately downstream of the stop codon (+4 position), also seem to determine the

read-through potential (C > U > A ≥ G) as well as nucleotides located one residue

upstream to the codon triplet (-1 position), where U seemed to induce the higher read-

through levels. The local sequence context around the PTC and its position within the

gene sequence where also shown to affect the induced read-through levels 26, 44, 45

.

The chemical structure of AG also seems to play a major role in their ability to

interfere with translation termination. In fact, not all AG are capable of inducing

suppression termination, and the explanation for these facts yet remains rather obscure.

In general, AG containing a 6'-OH group on their first ring (such as G418 and

Paromomycin) are superior to those enclosing an NH2 moiety at the same position 26, 46

.

These observations are usually subjected to one of the main differences between their

putative binding sites in eukaryotic vs. prokaryotic systems: the 1408 position (E. coli

numbering). It has been well documented that an A1408G mutation confers resistance to

AG, and that higher levels of resistance are usually observed towards AG containing an

amino moiety in their 6' position 47, 48

. The fact that eukaryotic ribosomes contain a

guanine residue in the corresponding position might explain the higher activity observed

for the 6'-OH derivatives. However, no structural evidence were presented to support

such biochemical observations.

Figure 1.4: A comparison of the A. normal

translation process leading to functional protein

production, B. translation which is interrupted by

a premature termination codon (PTC) leading to a

truncated non-functional protein production, and

C. translation process which was restored by a

read-through inducing compound such as an

aminoglycoside. Terminal codons and PTCs are

highlighted in yellow, aminoglycoside in green.

A.

B.

C.

Stop codon

Stop codon

Stop codon AG

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 24: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

12

1.4. The Read-Through Mechanism

In general, a read-through event is defined as the encoding of a PTC by a near

cognate aa-tRNA. Read-through events spontaneously occur in basal levels of less than

1% of translating PTC containing transcripts 49

. Therapeutic compounds inducing higher

read-through activity levels are believed to be able to enhance the possibility of near-

cognate aa-tRNAs to bind PTCs; therefore preferentially enhance their binding over the

binding of release factors (RFs), enabling the synthesis of full length transcripts. These

events can rarely occur on actual stop codons as extensively explained in Bedwell and

coworkers 49

, and are only limited to PTCs.

Recent progress in structural exploration of prokaryotic ribosomes deciphered the

mechanisms of which translation and translation termination occurs at the atomic level.

These findings altogether with the elucidation of AG mechanism of action in bacterial

cells might help in understanding how these therapeutic agents can induce read-through

of PTCs.

Overall, the termination of translation greatly differs from translating sense

codons. The translation of sense codons is performed by a careful selection of cognate aa-

tRNAs containing an appropriate anticodon sequence that can form Watson-Crick pairs

with all 3 codon nucleotides located in the mRNA template 50, 51

. Translation termination,

on the other hand, is encoded by only 3 consensus nucleotide triplets that can only be

recognized by proteins termed class I RFs 50-54

. Structural studies indicated an overall

structural similarity between the structures of RFs and aa-tRNAs. These findings

indicated that both factors occupy an ‘L’ shape conformation while bound to the

ribosome 50-54

. Moreover, both factors have been shown to be rather flexible. These

features are believed to enable the simultaneous interaction with both the ribosomal

decoding center - the A-site, located in the small ribosomal subunit, and the peptidyl

transferase center, located in the large ribosomal subunit.

At the A-site, both factors are able to recognize the relevant codon triplet (sense

codon/termination codon). tRNAs recognition is performed by tight monitoring of

specific hydrogen bonds between the anti-codon triplet located in the tRNA and the

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 25: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

13

codon triplet embedded in the mRNA template. RFs recognition is performed by specific

interactions of evolutionary conserved amino-acid (aa) motifs in the RF that tightly

interact with the 3 stop codon nucleotides. Upon recognition of the relevant codon

triplets, conformational changes occur in both the decoding center and the recognition

domain itself. These changes enable the direction of relevant domains of either the tRNA

or RF towards the peptidyl transferase center, where the synthesis of emerging

polypeptide chain occurs. These changes are irreversible and encompass the GTP to GDP

transfer of several extrinsic protein factors such as elongation factors (EFs) or additional

RFs (e.g. eRF3). In the peptidyl transferase center tRNAs catalyze the formation of

peptide bond between the relevant aa and the nascent polypeptide chain. RFs, on the

other hand, catalyze the hydrolysis of peptide chain, followed by its release, and the

beginning of ribosome recycling towards the next translational round (Figure 1.5).

At the atomic level, the above mentioned structural rearrangements upon codon

recognition greatly differ while comparing the two translational factors. The binding of a

cognate tRNA to the decoding center in bacterial ribosomes induces the flip out of the

two evolutionary conserved adenine residues, A1492 and A1493 (Figure 1.2). However,

structural studies on RF binding to bacterial ribosomes 50-54

, demonstrated that unlike in

the sense codon case, in the termination complex only A1492 flips out of H44, leaving

A1493 inside the helical core to establish stacking interactions with an adenine residue,

A1913, located at Helix69 (H69) in the large ribosomal subunit (Figure 1.5C). These

interactions are considered to be highly important for the proper termination of

translation process.

AG were long shown to stabilize the flipping out of the two conserved adenine

residues upon binding to bacterial ribosomes (Figure 1.3D). These interactions were

shown to affect ribosomal accuracy and lead to the production of mistranslated proteins

(Chapter 1.2). AG have also been shown to interfere with RF based peptide release and

induce read-through activity in prokaryotes quite long ago 20, 55

. However, only recent

kinetic indications have linked these interactions to RF inhibition in a competitive

manner 56

. Using the accumulated structural evidence presented here we are now capable

of understanding how such inhibition is possible at molecular levels. As indicated above,

AG binding to the A-site induces the flipping out of A1492 and A1493. The induced

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 26: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

14

conformation results in the steric clash of A1493 with RF, therefore, it might inhibit the

actual binding of RFs to the A-site (Figure 1.6). This conformation interferes with RF

binding to the A-site but also promotes the binding of a near cognate tRNAs (Figure

1.6); therefore enabling the resulting read-through activity. In addition, the flipping out of

A1493 prevents the stacking interactions with A1913 of H69 18

. Since these interactions

have been highlighted as important components of RF recognition by the ribosome, it

might add an additional explanation for their induced inhibition.

Figure 1.5 - Schematic view of translation (A) and translation termination (B,C).The left side in object C is

a representation of the 30S ribosomal subunit as obtained from the crystal structure coordinates (PDB ID:

2X9R), with mRNA highlighted in black; P-site and E-site tRNA molecules highlighted in green and blue,

respectively; and RF highlighted in red. The decoding site was highlighted in yellow and an enlarged

representation is presented in the right side of the figure. In the enlarged object RF is highlighted in red;

mRNA in gold; and the conserved adenine residue A1492 and A1493 in blue and purple, respectively.

A.

B.

C.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 27: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

15

Figure 1.6 - A. A molecular glance at the decoding site conformation upon RF binding to the bacterial A-

site. RF is highlighted in green; A1492 (blue) is flipping out from the helical core; A1493 (red) is directed

towards the inner part of the helical core stacking upon A1913 (purple) belonging to H69 at the large

ribosomal subunit (50S). Structures are derived from coordinations taken from PDB entry 2X9R. B.

superposition of the bacterial ribosome structure with the AG paromomycin (yellow sticks) bound to the A-

site (PDB ID: 3IBL) and RF bound to the bacterial ribosome (PDB ID: 2X9R). The structure demonstrate

the clashing of A1493 (red) with the Cα backbone of the RF (green), therefore suggests for a sterical

hindrance of RF to the A-site upon the induced conformation when AG is bound to the bacterial A-site.

These recent findings highlight the possibility that enhanced read-through events

occur due to steric interference of ribosomes with RFs, limiting their binding affinities to

stop codons while enhancing the binding of near-cognate tRNAs to PTCs. These findings

have only been partially demonstrated experimentally in bacteria, however, up to this

date, no such information regarding the read-through mechanism in eukaryotes is yet

available.

A. B.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 28: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

16

1.5 Aminoglycosides Activity against Parasitic Protozoa - Leishmania

AG activities in eukaryotes were extensively investigated over the last few

decades and as such it has been found that some AG could be used as highly potent

agents for the treatment of infections caused by parasitic protozoa such as

trypanosomiasis, giardiasis, amoebiasis and leishmaniasis 57-61

. Very recently,

Paromomycin, a clinically approved AG for the treatment of bacterial infections was

suggested as an alternative treatment of Leishmaniasis.

Leishmaniasis is a spectrum of diseases caused by parasitic protozoa belonging to

the genus Leishmania. It affects millions of people worldwide, appearing mainly in

tropical and subtropical areas 61

. The parasite is transmitted by sandflies, and depending

on the type of disease can be fatal if untreated. The current state-of-the-art in treating

leishmaniasis is based on chemotherapy using a limited array of drugs such as antimony

containing agents, amphotericin B, and Miltefosine 61

. However, due to the emergence of

pronounced parasite drug resistance in some regions, relatively high costs, and/or severe

toxic effects, there has been an extensive search over the last few years for new

therapeutic agents. As such, Paromomycin has been highlighted as a promising candidate

which is now tested in various clinical trials worldwide 61

. Paromomycin was found to be

effective against a wide range of Leishmania parasites and was suggested for the

treatment of both cutaneous and visceral leishmaniasis, which is the fatal form of this

disease 62

. Paromomycin is already being clinically used as the major component of a

topical ointment (Leishcutan) used to treat cutaneous leishmaniasis, and it is registered in

India and Nepal for the treatment of visceral leishmaniasis. Clinical trials using

Paromomycin in combination with other anti-leishmanial drugs are underway in order to

prevent development of parasite resistance 63

.

A very recent paper has demonstrated Paromomycin's ability to inhibit

leishmanial ribosome synthesis in-vitro, and cause translational misreading events in-

vitro. However, despite the great potential in the use of AG as anti-protozoal therapeutic

agents, very little information is available regarding their molecular mechanisms of

action in eukaryotes, and to our knowledge, no comprehensive study has been designed

to test the susceptibility of such parasites to various AG derivatives.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 29: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

17

1.6 Aminoglycosides Toxicity

Despite the promising results indicating the great potential of using AG to treat

eukaryote related disorders, their use for these therapeutic purposes is quite restricted

nowadays. These limitations are mainly due to the relatively high toxicity associated with

their prolonged administration in eukaryotic systems. The mechanisms of which AG

induce their toxic effects in eukaryotes are not fully deciphered yet; the current

information in this field is very little, and is mainly attributed to their positively charged

nature that makes them prone to interact with various negatively charged cellular

components such as phospholipids, phospholipases and various metal ions. These

interactions are believed to eventually lead to the generation of reactive oxygen species

(ROS) 64

which are known to enhance cell toxicity.

AG attribution to the inhibition of eukaryotic translation machinery is also

considered to play a major part in their induced toxicity. However, recent publications

highlighted the mitoribosome as a main target responsible for AG toxic effects. These

papers demonstrated the ability of some natural derivatives to inhibit mitochondrial

protein synthesis; apparently, due to the relatively high similarity the mitochondrial

ribosome shares with their primary target site, the bacterial ribosome 65, 66

. Bacterial and

mitochondrial A-sites 2-dimensional structures are presented in Figure 1.7.

Over the last few years, many efforts have been directed in an attempt to

overcome the toxicity associated with AG administration. Those included co-

administration with a variety of compounds including several antioxidants such as

vitamin E 67

and salicylic acid 68-71

, iron chelators 70, 72

, and some negatively charged

agents such as poly-L-aspartate 73, 74

and daptomycin 75, 76

, which are considered to limit

their unspecific interactions with

negatively charged cellular components.

Recent papers have suggested various

synthetic derivatives with a relatively

reduced toxicity when compared to the

natural compounds.

A. B.

Figure 1.7: Bacterial (A) vs. Human

mithochondrial A-site (B).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 30: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

18

A more recent paper demonstrated the ability to diminish the induced toxicity by the

development of synthetic moieties that selectively target the cytoplasmic eukaryotic

ribosome, avoiding mitochondrial inhibition. These derivatives will further be discussed

in Chapter 1.7.

1.7 Challenges in the Design of Novel Drugs Targeting the Eukaryotic Ribosome

Synthetic derivatives caring an aminoglycosidic scaffold have been extensively

investigated over the last few decades as potential therapeutic agents to be used for the

treatment of general bacterial infections. These efforts resulted in the development of

improved derivatives with reduced toxicity, and enhanced ability to delay the emergence

of resistance among bacterial species. These compounds were extensively investigated in

both biochemical and structural manners, and along with the molecular understanding of

AG mechanisms of action and resistance in bacterial species, opened a new avenue in the

field of antibacterial therapy. However, due to the limited information regarding their

mechanism of action in eukaryotes as well as the higher complexity of these systems,

when compared to the prokaryotic ones, only a few novel semi-synthetic derivatives have

been suggested for the treatment of eukaryote related diseases (Figure 1.8).

1.7.1 Development of Read-Through Inducers

In 2006, Lorson and coworkers 77

demonstrated the ability of 6 semi-synthetic

Neamine and Kanamycin derivatives, such as compound 1 (Figure 1.8), also named

TC007, to promote read-through of PTC in survival motor neuron-1 (SMN) in fibroblasts

derived from SMA patients. These agents have shown to induce an up to a 30 fold

increase in normal protein production compared to the untreated baseline in a dose

dependent manner. Some compounds have shown to induce better activity than the well

documented histone deacetylase inhibitor, valoparic acid (VPA), which is known to

induce SMN expression in mutant cells 78, 79

. TC007 has later been reported to induce

beneficial effects on muscle fiber size and gross motor function in an SMN mice model

77, 80. However, the synthetic derivatives tested within this assay were initially designed

as antibacterial agents 81

therefore contained an amino group in their 6' position.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 31: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

19

Unfortunately, no direct comparison to the natural aminoglycosidic scaffold has been

reported in this manuscript. Comparison of the reported results with earlier work

presented by the same authors indicated similar ability of natural derivatives such as

Tobramycin and Amikacin to induce read-through in SMA patient fibroblasts 40

.

In 2006, Baasov and coworkers first reported on the development of the semi-

synthetic Paromomycin derivative, NB30 (compound 2) 82

(Figure 1.8) to be used as a

prototype read-through inducer. NB30 has been shown to induce PTC suppression in

vitro and ex-vivo against DNA fragments that mimic genes with disease causing nonsense

mutations such as DMD, HS, USH and CF 82, 83

, and recently on mutant retinal explants

derived from mice 84

. Comparison of the obtained results with those obtained for natural

derivatives such as G418, Paromomycin and Gentamicin, indicated lower activity of the

synthetic derivative. However, cell toxicity assays 83

, altogether with ototoxicity assays

performed in cochlear explants and acute toxicity experiments in mice 85

indicated

reduced toxicity of the synthetic derivative when compared to the natural inducers.

Encouraged by these results these researchers further reported in 2009 the

development of the new and improved Paromomycin based synthetic derivate, NB54

(compound 3), that contained an (s)-4-amino-2-hydroxybutanoyl (AHB) moiety at N-1

position (Figure 1.8) 85

. The new structure was designed to have lower toxicity values

comparing to its prototype based on previously documented results indicating decreased

lethal toxicity values (LD50) in mice treated with natural derivatives containing AHB in

their N-1 positions 86

. The resulting structure indeed exhibited a much reduced toxicity

values when compared to its prototype, NB30 85

, and has also been shown to induce an

enhanced read-through activity in-vitro 85

, ex-vivo 33, 34

and in-vivo 87

. NB54 activity

exceeded Paromomycin and Gentamicin activities.

Approximately one year later, the group reported the development of two new

synthetic derivatives in which the first two rings of G418 were used as the

aminoglycosidic scaffold (Figure 1.8; compounds 4,5, designated NB74 and NB84) 88

.

These derivatives were designed to contain a 6'-(R) methyl that we hypothesized to act as

a pharmacophore in G418, but nevertheless, did not contain the garosamine 3rd

ring,

which has been shown to enhance toxicity in our previous studies. Evaluation of the

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 32: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

20

resulting compounds indicated better read-through activity in vitro and ex-vivo, along

with significantly reduced cell toxicity. Compound 5 has later been shown to induce read-

through activity in an HS mice model 89

.

Continuous research in our lab has recently led to the development of new

generations of synthetic derivatives which have not only been shown to be potent read-

through inducers with relatively low toxicity, but also to be highly selective towards the

eukaryotic ribosome (Figure 1.8; compounds 6-13, NB118-119, NB 122-125 and NB

127-128) 90, 91

. These derivatives were introduced with a new pharmacophore - 5'' (R/S)

methyl. Compounds 12 and 13 were shown to have read-through activity similar to G418

in both in-vitro and ex-vivo systems, but exhibited a much reduced cell toxicity.

The above mentioned progress in designing synthetic derivatives with potent

read-through potential and low toxicity emerged from a very careful inspection of the

structural elements which are highlighted as important for the biological activity and

toxicity of AG. These recent results are indeed very encouraging, but unfortunately,

rational design of synthetic AG is still far from being well established mainly due to the

lack of detailed information in regarding to the mechanisms involved in AG activity and

toxicity in eukaryotic systems. Our recent publications demonstrated the importance of

high selectivity towards the eukaryotic cytoplasmic ribosomes for the development of

therapeutic agents with reduced toxicity 91

. However, compounds that exhibit an

enhanced selectivity towards eukaryotic systems do not always exceed better read-

through activity. The natural AG Apramycin and the synthetic derivative NB33 (Figure

1.9) where shown to exhibit a rather high selectivity towards eukaryotic systems 92

;

nevertheless, their measured read-through activity appeared to be rather negligible 92

.

Over the last several years a few more AG based synthetic derivatives have been

evaluated as selective potent binders of eukaryotic systems 92

. However, the estimation of

their read-through abilities is yet to be determined.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 33: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

21

O

NH2

NH2

OH

O

O

NH2

HOHO

H2N

6'

5

4

O

HOH2NHO

HO

1 (TC007)

O

NH2

NHR

OH

O

O

OH

HOHO

H2N

6'

5

4

2 (NB30): R=H

O

HO OH

H2N

3 (NB54): R=AHB

5''

O

OH

NH2

AHB =

O

NH2

NHR

OH

O

O

OH

HOHO

H2N

6'

5

4

4 (NB74): R=H

O

HO OH

H2N

5 (NB84): R=AHB

5''

Me

O

NH2

NHR

OH

O

O

OH

HOHO

H2N

6'

5

4

6 (NB119): R=H; *(R)

O

HO OH

NH2

7 (NB123): R=AHB; *(R)

5''

Me *

8 (NB118): R=H; *(S) 9 (NB122): R=AHB; *(S)

O

NH2

NHR

OH

O

O

OH

HOHO

H2N

6'

5

4

10 (NB125): R=H; *(R)

O

HO OH

NH2

11 (NB128): R=AHB; *(R)

5''

Me *

13 (NB127): R=AHB; *(S)12 (NB124): R=H; *(S)

Me

OH

H2NH2N

HO

O

O

HO

OHO

NH2

HO

NH2

NH2

OH

O

O

OH

HOO

H2N

NB33

ApramycinHO

NH2

NH2

OH

O

O

OH

H2N

OO

O

HO

OH

H2NHO HN

Figure 1.8: Structures of novel semi-synthetic AG that induce translational read-through

of premature stop codons (compounds 1-12) and compounds exhibiting some high

selectivity towards eukaryotic systems but lacking any read-through activity (Apramycin

and NB33).

1.7.2 Development of Anti-Protozoal Derivatives

AG anti-protozoal activity has been discovered in the 70s 60

, however, only in

recent years Paromomycin has been suggested clinically for the treatment of such

protozoa related infections 61

. As a result, only recently, there have been evidence for the

development of new and improved derivatives for the treatment of leishmaniasis,

giardiasis and trypanosomiasis 59

. In these recent publications, Hanessian and coworkers

describe the preparation and evaluation of two synthetic therapeutic derivatives that

contain an unsaturated ring I (compound 1a and 1b, Figure 1.9). The synthetic

derivatives are 6'-OH derivatizations of Sisomycin, which is an unsaturated natural AG,

exhibiting some excellent antibacterial activities with relatively low toxicity values 59

.

Derivative 1a, also known as 6'-OH Sisomycin, is nearly identical to Sisomycin,

containing only a single modification in regards to Sisomycin (6'-OH group instead of a

6'-NH2 group). Derivative 1b, also known as Paromo-Siso-hybrid, is a simple

hybridization of the 6'-OH Sisomycin's rings I and II with rings III and IV of

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 34: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

22

Paromomycin (Figure 1.9). The presented in-vitro susceptibility assays, performed in a

few strains of pathogenic parasites, indicated IC50 values for derivative 1a at the mid-low

μM range 59

. These values were around 1 order of magnitude lower than the ones

observed for Paromomycin, which is currently under clinical trials worldwide. The IC50

values indicated for compound 1b, were similar to those reported for paromomycin. Cell

toxicity values (LC50) performed in rat skeletal muscle (L6) cells indicated similar

toxicity values for both synthetic derivatives and Paromomycin 59

.

IV

II

III

I

O

ONH2

OH

OH

H2N

OHO

O

OH

NH2

NH2

OH

O

O

OH

H2N

6'

5

4I

HO

NH2

NH2

O

O

OOH

H2N

OHO

HN OH

II

III

6'

4

6

1a (6'-Hydroxysisomycin) 1b (Paromo-siso hybrid)

I

HO

NH2

NH2

O

O

ONH2

H2N

OHO

HN OH

II

III

6'

4

6

Sisomycin

Figure 1.9: Structures of novel semi-synthetic AG exhibiting some anti-protozoal properties.

1.8 Aminoglycosides and the Eukaryotic Ribosome - Current Knowledge and

Alternative Model Systems

The elucidation of x-ray crystal structures of bacterial ribosomes at the turn of the

century enabled a "ribosomal renaissance", allowing relationships between structure and

function to be studied at the atomic level 93-96

. However, the structural information

regarding eukaryotic ribosomes was limited until very recently to medium resolution cryo-

EM single particle reconstructions, due to the more complex nature of eukaryotic systems

97, 98. Recent structural work revealed the three dimensional structures of the 80S yeast

ribosome 99, 100

and the 40S, 60S subunits of the protozoa Tetrahymena thermophila 101, 102

at relatively high resolutions. This recent structural information opens a variety of

opportunities underlying the variance in eukaryotic vs. prokaryotic translation mechanisms

together with unlimited possibilities to explore the interactions of small molecules, such as

AG, with eukaryotic ribosomes. Nevertheless, one mustn't forget that this field is still very

young and the study of complex molecules such as ribosomes still remains very

complicated.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 35: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

23

As an alternative path, over the last several years attempts to provide a molecular

basis for AG activity in mammalian ribosomes have resulted in the development of

various eukaryotic rRNA constructs (Figure 1.10) 103-109

, that mimic putative AG binding

sites in eukaryotic systems. Such constructs (which hence will be called models)

contained only a minimal A-site stabilized with additional non-natural Watson-Crick

pairs. Similar constructs were extensively used for the exploration of AG activity in

prokaryotes and were proven to be highly efficient in mimicking the bacterial A-site.

Superimposition of crystal structures of Paromomycin bound to the 70S apparatus and a

minimal bacterial A-site model is presented in Figure 1.11. The demonstration of a rather

high similarity between the obtained conformation in the minimal model and the one

obtained upon binding to the whole ribosomal apparatus altogether with the simplicity of

using such minimal models for crystallization experiments resulted in a rather extensive

use of such models for the exploration of AG activities in prokaryotes and the rational

development of new and improved antibacterial derivatives 4, 110-114

. The suggested

eukaryotic constructs were based on the high level of rRNA binding site conservation

among prokaryotic and eukaryotic systems (Figure 1.10 A,B); therefore they are believed

to properly represent the ribosomal eukaryotic binding sites.

The resulting constructs were extensively used as models to explore the nature of

various AG interactions with their eukaryotic binding sites. Investigations of the physical

properties involving AG interactions with their putative binding sites have been

thoroughly demonstrated by the use of methods such as isothermal titration calorimetry

(ITC) 107, 115, 116

, UV melting experiments 107, 115, 116

, circular dichroism (CD) 115, 116

,

nuclear magnetic resonance (NMR) 105

, and various fluorescence based studies including

fluorescence resonance energy transfer (FRET) 107-109

. The obtained results indicated

binding affinities at the millimolar range, approximately 3 orders of magnitude higher than

their binding to the corresponding prokaryotic binding sites. These values are well

correlated with in-vitro, ex-vivo and in-vivo studies that evaluated AG ability to induce

read-through activity in eukaryotic systems 82, 85, 90-92, 117

. The obtained affinity gap has

been linked to the minor sequential differences between the eukaryotic and the prokaryotic

binding sites which are located at positions that are considered to be important for AG

binding, such as A1408G and G1491A (Figure 1.10).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 36: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

24

Three of the suggested models were exploited for structural exploration and their

three dimensional structures have been determined by X-ray crystallography 92, 103, 104, 118

or NMR 105, 106, 119

. The structure of a hairpin model suggested by Puglisi and coworkers

has been solved in complex with Paromomycin by NMR spectroscopy 105

(Figure 1.10C).

The global RNA structure in the resulting model was rather similar to the one obtained

from the NMR structure of a corresponding bacterial A-site model in complex with

Paromomycin 120

. However, in the eukaryotic model, the paromomycin binding pocket

was slightly shifted, and its binding did not induce the flipping out of the two conserved

adenine resides as usually observed in the corresponding prokaryotic binding site (Figure

1.12). Nevertheless, while analyzing these data, two important issues should be taken

under consideration: i. the mobility of A1492 and A1493 in the bacterial NMR model was

also limited in comparison to the one observed in X-ray experiments 120

. ii. the RNA

construct used in this study corresponds to the Tetrahymena thermophila decoding site,

and therefore is quite remote from the sequence of the Homo sapiens putative binding site.

In 2006 an RNA construct, that contained two minimal eukaryotic A-site cores

separated by four G=C pairs, was suggested by Westhof and coworkers 121

(Figure

1.10G). The construct was based on a similar previous prokaryotic model that has been

shown to properly mimic the AG prokaryotic binding site and corresponded to the Homo

sapiens A-site sequence 113

. The suggested model has been used for the exploration of

both A-site structure and AG mechanism of action in eukaryotic systems. X-ray structure

determination of a "ligand free" construct indicated two different conformations of the two

binding sites present within the model. These two conformations were suggested to

correspond to an 'ON' state, with the two conserved adenine residues A1492 and A1493

fully bulged-out, and an 'OFF' state, where A1491 fully bulged-out and A1493 directed

toward the helical core (Figure 1.12B-D). The same 'OFF' state conformation was later

found in a crystal structure of a different RNA construct that was used for the exploration

of AG interaction with their putative binding site 104

(Figure 1.10F).

Recently, Westhof and co-workers solved the crystal structures of their eukaryotic

A-site model in the presence of two AG with high selectivity toward the eukaryotic

ribosome 92, 118

: the natural AG Apramycin and the synthetic derivative NB33 that has

been developed in our lab (Figure 1.7). Interestingly, these AG have been shown to

stabilize the 'OFF' state conformation (Figure 1.12E,F). Since these two agents have also

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 37: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

25

been shown to be effective inhibitors of the eukaryotic ribosome and lack any read-

through activity 92

these recent achievements may provide indirect evidence for the

similarity of AG mechanism of action in eukaryotic systems to their mechanism of action

in the bacterial ribosome. However, to date, no crystal structure has been solved in the

presence of an AG with read-through ability, and direct evidence is currently unavailable.

Figure 1.10: Two dimensional representation of the bacterial (A.) vs. eukaryotic cytoplasmic (B.)

decoding sites (A-sites). The two conserved adenine residues A1492 and A1493 are highlighted in blue and

red, respectively. G/A1491 in green, and A/G1408 in orange. C-G. Synthetic models of the eukaryotic A-

site used for the exploration of aminoglycosides interactions with their putative binding site. A-site

sequence is highlighted in a red dotted square. Capital letters represent a natural sequence. Small letters

represent synthetic sequences added to stabilize the model or generate a loop. The two hairpin models (C.

and D.) were suggested by Puglisi 105, 106

, Tor 108

and Pilch 107

. Model C. was used for NMR experiments;

Model D. was used for fluorescence spectroscopy experiments. The position marked in * was tagged in a

fluorescence acceptor for FRET experiments. Model E. was suggested by Vourloumis 109

and was also

used for fluorescence spectroscopy exploration. Apart from FRET experiments performed by Tor and

collogues A1492-3 were replaced by 2-aminopurine (2-AP) residues in the fluorescence experiments.

Models F. and G. were suggested by Hermann 104

and Westhof 103

, and were used for X-ray

crystallography.

A. B. C. D. E. F. G.

Bacterial

E. coli Human

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 38: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

26

Figure 1.12: The eukaryotic A-site models used for the structural exploration of aminoglycoside binding site. A.

NMR structure of the hairpin model suggested by Puglisi and coworkers in complex with paromomycin (PDB

ID: 1FYP). Paromomycin is shown in green ball-and-sticks representation, two conserved adenins 1492, 1493

are highlighted in blue and red respectively. B. X-ray crystal structure of the RNA duplex containing two

minimal eukaryotic A-sites suggested by Westhof and colleagues (PDB ID: 2FQN). Two different

conformational states were demonstrated within the same construct: C. 'ON' state conformation with adenine

residues A1492 (blue) and A1493 (red) fully bulged-out, and A1491(green) directed toward the helical core. D.

'OFF' state conformation with A1491 (green) and A1493 (red) bulged out and A1492 (blue) directed inside the

helical core. E,F. 'OFF' state conformation induced by apramycin (E.) (PDB ID: 2J5K); and the synthetic

derivative NB33 (F.) (PDB ID: 2O3V).

A. B. C.

D.

E.

F.

A 1493

A 1492

Paromomycin

Figure 1.11: Superposition of Paromomycin

bound to a minimal A-site model (blue, PDB ID:

1J7T) and to the 70S ribosome apparatus (orange,

PDB ID: 1IBL). RMSD 0.69Å.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 39: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

27

2. Research Goals and Perspectives

AG hold great potential to serve as therapeutic agents for the treatment of PTC

induced genetic disorders and parasite related infections. This potential has been already

demonstrated dozens of years ago, and has recently been dulled due to their induced toxic

effects in eukaryotic species. Recent efforts in drug design have resulted in the

development of new and improved derivatives that might overcome these drawbacks and

makes them feasible candidates for such therapeutic purposes. These encouraging recent

achievements have also demonstrated the current need in understanding the molecular

mechanisms by which AG exert their biological activity in eukaryotic cells for rational

development of new therapeutic agents.

To date, no structures of the eukaryotic A-site in complex with any of the AG

exhibiting the relevant biological activity are yet available, and the elucidation of the read-

through mechanism in eukaryotic systems as well as killing mechanisms in parasites are

yet to be discovered. Nevertheless, the relatively high similarity of AG binding sites in

bacterial ribosomes to their putative binding sites in the eukaryotic ribosomes, altogether

with the recent development in eukaryotic ribosome structures makes it likely to assume

that these mechanisms share great similarity. However, further detailed investigations are

needed in order to establish their mechanism of action in eukaryotic system making it

practicable for rational drug design.

The main goals of this study were to ascertain the molecular mechanisms of AG-

induced read-through in human cells as well as to elucidate their mechanisms of action in

eukaryotic parasites. To explore such mechanisms we chose to use X-ray crystallography

as a tool to investigate the unique binding pattern of AG with their putative binding sites

in eukaryotes. The structural studies were correlated with biochemical investigations

performed in-vitro and ex-vivo in order to measure the biological activities of the tested

compounds in the relevant eukaryotic systems, as well as to the chemical synthesis of new

and improved therapeutic derivatives. We truly believe that such interdisciplinary studies

performed under the same roof can give rise to the rational development and improvement

of AG as therapeutics for eukaryote related diseases.

In addition to the structural studies, the present study was aimed at developing a

highly sensitive assay for the determination and quantification of AG which are highly

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 40: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

28

selective to eukaryotic systems in biological samples. Such assay will be of a great use in

the further development of these therapeutic compounds, allowing us to monitor their

activities in-vivo. To achieve these goals we chose to develop some highly sensitive

polyclonal antibodies (Abs) that will be able to detect some highly conserved epitopes

present in the majority of therapeutic compounds. Such Abs could be used for the

development of a highly sensitive immunoassay for the determination of AG in various

biological matrices. Such tools could also be exploited for testing some additional

mechanistic aspects of AG activities in eukaryotes such as their permeability

characteristics and could be of a great help improving their therapeutic indexes.

The specific goals of the present study included: (a) Co-crystallization and

determination of the three-dimensional structures of natural and synthetic lead AG with

RNA fragments that reproduce their eukaryotic putative binding sites (A-site); (b)

determination of the molecular basis that governs the AG selectivity towards bacterial

versus eukaryotic ribosomal machineries. (c) Performance of an in-depth chemical

analysis of the modes of structural recognition to elucidate the mechanism by which AG

induce stop-codon read-through and anti-protozoal activities. (d) Performance of docking

and modeling experiments using the resulting molecular coordinates for the rational

design of new derivatives. (e) Preparation of highly selective polyclonal Abs that will be

able to recognize 6'-OH AG derivatives. (f) Development and employment of a highly

selective immunoassay for the detection of such derivatives in various biological matrices.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 41: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

29

3. Materials and Methods

3.1 Materials

Geneticin (G418) and Apramycin were purchased from Apollo Scientific Limited,

UK as sulfate salts. Gentamicin sulfate was purchased from Molekula, UK. All the

synthetic derivatives including neamine, paromamine, NB82, NB74, NB84, NB122,

NB123, NB124 and NB125 were prepared as described previously by Baasov and

coworkers 85, 88, 90, 91

and were used as their sulfate salts. All other chemicals and

biochemicals, unless otherwise stated, were obtained from Merck or Sigma.

3.2. General Techniques

3.2.1 X-ray Analysis

X-ray measurements were performed using synchrotron radiation at a wave length

of ~0.9Å, or by using Rigaku Raxis IV++ and NarµX X-ray System machineries with a

fixed wavelength of 1.54Å (Tel-Aviv and Ben-Gurion Universities, respectively).

Synchrotron radiation experiments were performed either in the Swiss Light Source

(SLS) facility located at the Paul Scherrer Institut (PSI) in Villigen, Switzerland (PXI) or

at the European Synchrotron Radiation Facility (ESRF) located in Grenoble, France

(ID14-1, ID14-4, ID23-1, ID23-2 and ID29). Measurements were performed on frozen

crystal samples that were mounted on nylon cryo-loops (Hampton research) prior to flash

freezing in liquid nitrogen.

3.2.2 NMR, MS and TLC

1HNMR (Nuclear Magnetic Resonance) spectra was recorded on a Bruker Avance™ 500

spectrometer at 500 MHz. Chemical shifts reported (in ppm) were relative to the internal

standard Me4Si (δ = 0.0) with CDCl3 as the solvent. 13

CNMR spectra was recorded on a

Bruker Avance™ 500 spectrometer at 125.8 MHz, and the chemical shifts reported (in

ppm) were relative to the residual solvent signal for CDCl3 (δ = 77.00). Mass spectra

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 42: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

30

(MS) analysis was obtained on a Bruker Daltonix Apex 3 mass spectrometer under

electrospray ionization (ESI). Reactions were monitored by Thin Layer Chromatography

(TLC) on Silica Gel 60 F254 (0.25 mm, Merck), and spots were visualized by charring

with a yellow solution containing (NH4)Mo7O24•4H2O (120 g) and (NH4)2Ce(NO3)6 (5 g)

in 10% H2SO4 (800 mL). Flash column chromatography was performed on Silica Gel 60

(70–230 mesh).

3.3 RNA Purification and Crystallization

3.3.1 RNA Purification

RNA constructs corresponding to the human and leishmanial A-sites were

chemically synthesized by Dharmacon (Boulder, CO, USA). RNA sequences used were

as follows: Human A-site used for the double A-site model experiments was composed of

two identical RNA sequences: 5'-UUGCGUCGCUCCGGAAAAGUCGC-3'; Human A-

site used for the single A-site experiments was composed of two different RNA

sequences: (a) 5'-GGGCGUCGCUAGUACCG-3', (b) 5'-GGUACUAAAAGUCGCCCC-

3'. Leishmanial double A-site model was composed of a single RNA sequence

corresponding to the leishmanial A-site: 5'-UUGCGUCGUUCCGGAAAAGUCGC-3'.

In order to avoid RNA degradation during shipment the RNA oligos arrive with

an acid-labile orthoester protecting group on the 2'-hydroxyl (2’ACE). Prior to

crystallization these protection groups had been removed in order to “activate” the RNA

oligos. Deprotection procedure has been performed at pH 3.8, 60ºC for 30 minutes.

Following deprotection the oligo RNAs were purified by anion exchange purification

under denaturating conditions (8M urea, 60ºC) on AKTA FPLC device connected to a

UV detector. Elution was performed by gradually increasing the NaClO4 concentration

within the buffer solution (1-400 mM). Fractions of 1 ml were automatically collected

and the purity level of RNA containing samples has been analyzed by analytical PAGE

(Figure 4.2). Samples with high purity were pooled for desalting. Desalting was

performed by using Sep-Pac C-18 column (Waters) that was preconditioned by a wash

with 10 ml of absolute ethanol, followed by a wash with 10 ml of MilliQ water. RNA

samples were loaded on the columns, which were then washed with 10 ml MilliQ water.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 43: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

31

Elution was carried out with 5 ml of 80% ACN. The samples underwent vacuum

evaporation and dissolved in MilliQ water to final concentration of 4mM. The RNA

concentration was determined by spectroscopic measurement of absorption at 260nm.

3.3.2 RNA Annealing

In order to avoid heterogeneity in the RNA constructs' conformations, purification

was followed by additional steps of denaturation (at 90˚C) and annealing (by slow

cooling to 37˚C, ~2h). These stages ensure the formation of thermodynamically stable

RNA constructs with maximal base-pairing. In experiments were ligand interactions with

the RNA construct were examined, an additional step of ligand introduction was added.

Generally, the ligand will be mixed with the RNA constructs at 37˚C for 10 min in

various ligand:RNA ratios prior to crystallization. The RNA solution used in the X-ray

experimentation was usually composed of 2 mM RNA in 100 mM sodium cacodylate pH

5.0-7.5 and 25 mM NaCl.

3.3.3 Crystallization

Crystallization experiments were performed at 20°C using the hanging-drop vapor

diffusion method. Droplets were prepared by mixing 1 μL of RNA/AG solutions and 1

μL of crystallization solutions as indicated in Tables I, II (Appendix). In general, most

crystallization experiments were performed in 24 wells plates (Hampton reaserch) that

were sealed with appropriate rounded siliconized glasses (Hampton research). In most

experiments 500 μL of 40% 2-4-methyl-pentadiol (MPD) was used as a reservoir

solution. Crystals usually emerged after 4-6 days.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 44: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

32

3.4. RNA-models: Crystal Handling, Data Collection, Structure Determination and

Refinement

3.4.1 Standard Crystallization Procedures and Data Handling

Normally, crystals were soaked in 40% MPD and flash frozen in liquid nitrogen.

Frozen crystals were transferred to the SLS or ESRF where X-ray data were collected.

Data processing has been performed with XDS 122

or Mosflm 123

. Initial phases were

determined by molecular replacement using Phaser MR, CCP4 suite 124

, using the

coordinates of previously solved X-ray structures of similar models as a search model.

The molecular structures were constructed and manipulated with COOT 125

. Data

refinement has been performed using Refmac5, (CCP4 suite), Phenix Refine 126

, and

CNS 127, 128

. Graphical representations were made using PyMOL 129

. The atomic

coordinates and structure factors of G418 and Apramycin A-site complexes with the

leishmanial binding site were deposited in the Protein Data Bank (PDB) with the

accession codes 4K32 and 4K31, respectively.

3.4.2 Additional Crystallization Techniques (Seeding, Soaking)

Since some of our crystallization trials resulted in poorly diffracting crystals or

structures exhibiting only a mere indication of ligands presence some of our

crystallization experiments and crystal handling techniques were performed using some

additional crystallization techniques such as seeding and soaking. Seeding and soaking

are highly acceptable methodologies in crystallography that are being used extensively in

crystallographic researches 130

.

For crystals that diffracted well to high resolution but did not exhibit a strong

indication for ligand's presence we applied the soaking technique where we added high

concentrations of the relevant ligand to the cryo-solution prior to freezing the crystal in

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 45: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

33

liquid nitrogen. In these experiments we soaked the crystals for 2-24 hours in a 40%

MPD solution containing high ligand concentrations (10 mM and up) altogether with

polyamines and salts compositions at similar concentrations to the ones used in the

crystallization conditions.

For small crystals that gave a poor diffraction we used both macro- and micro-

seeding techniques. In our macro-seeding experiments we transferred the small occurring

crystals as is to a droplet containing 40% MPD for a few minutes (this procedure has

been done in order to wash the outer shell of the crystals) and then transferred them to a

new crystallization droplet containing RNA, AG and all relevant crystallization

conditions. The droplets have been equilibrated over a 40% MPD reservoir solution at

20˚C. Our micro-seeding experiments included the use of a cat mustache hair as a rough

micro surface that was transferred over the crystals a few times (this action should have

resulted in the clinging of micro crystals on the hair's jAG; transferring the hair in a

freshly prepared crystallization solution containing all relevant reagents introduced the

newly prepared droplet with the micro crystals that could be used as nucleation surfaces

for the emergence of new crystals.

3.5 Yeast 80S Ribosomes Crystallization and Crystal Structure Determination

Our crystallization trials of yeast 80S ribosomes complexes with various AG were

conducted in Prof. Marat Yusupov's lab in Strasbourg University, France. The

purification process of 80S ribosomes from JD1370 yeasts has been performed as

previously described by Yusupov and coworkers 100

. In brief, yeast cells were grown in

flasks to an OD600 of 1.45 in YPD at 30˚C. Cells were pelleted by centrifugation, re-

suspended with YP and incubated for 10 min at 30˚. Cells were incubated on ice and

were precipitated and washed three times in a buffer containing 30mM Hepes-K pH 7.5,

50 mM KCl, 10 mM MgCl2, 8.5% mannitol, 2mM DTT, 0.5 mM EDTA). For 4.5 gr of

cells, the pellet was resuspended in 6.5 ml of the same buffer solution and supplemented

with additional 600 μL from a solution of one complete protease inhibitor tablet (without

EDTA, Roche) dissolved in 2 ml buffer solution, 100 μL RNasin (Promega), 120 μL

Pefablock 100 mM and 56 μL freshly prepared Naheparin 100 mg/ml. Lysis has been

performed by adding 425-600 μm glass-beads (Sigma) into the the suspended tube and

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 46: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

34

maintaining 5 cycles of 1 min vortexing followed by equal time of incubation at 4˚C.

Beads were removed by short centrifugation (20,000g * 2 min) and the lysate was further

clarified by a longer centrifugation (31,000g * 9 min). PEG 20,000 was then added from

a 30% w/v stock (Hampton Research) to a final concentration of 4.5% w/v and the

solution was left to stand for 5 minutes on ice. The solution was clarified by

centrifugation (20,000g * 5 min) and the supernatant was decanted to a new tube. The

KCl concentration was then adjusted to 130 mM. After 5 min on ice PEG 20,000

concentrations were adjusted to 8.5% and the solution was left to stand for 10 min on ice.

Ribosomes were precipitated (17,500g * 10 min), the supernatant was discarded and

residual solution was removed by a short spin of the pellet (14,500g * 1 min). Ribosomes

were suspended (6.5-7 mg/ml) in a buffer solution similar to the buffer solution used for

cells lysis of which the KCl concentration was adjusted to 150 mM and supplemented

with protease inhibitors and heparin. Ribosomes were further purified by a 15-30%

sucrose gradient in buffer A (20 mM Hepes-K pH 7.5, 120 mM KCl, 8.3 mM MgCl2, 2

mM DTT, 0.3 mM EDTA) using the SW28 rotor (18,000 rpm * 15h). The appropriate

fractions were collected, KCl and MgCl2 concentrations were adjusted to 150 mM and 10

mM respectively, PEG 20% was then added to a final concentration of 7% w/v and the

solution was left to stand 10 min on ice. Ribosomes were precipitated (17,500g * 10

min), the supernatant was discarded and residual solution was removed by a short spin of

the pellet (14,500g * 1 min). Ribosomes were suspended (20 mg/ml) in buffer G (10 mM

Hepes-K pH 7.5, 50 mM KOAc, 10 mM NH4Cl, 2 mM DTT, 5 mM Mg(OAc)2).

Prior to crystallization, the ribosome stock solution was filtered (0.22μm

centrifugal filters, Millipore) and a ribosome solution was prepared containing 5 mg/ml

ribosomes, 2.5 mM Hepes-K pH 7.5, 2.5 mM NH4Cl, 3.33 mM Mg(OAc)2, 1.6 mM DTT,

0.055 mM EDTA, 2.8 mM Deoxy Big Chap, 40 mM KOAc, 5.5 mM NH4OAc, 5.5 mM

Tris-Acetate pH 7.0. The ribosome solution was incubated in the presence or absence of

AG at 30˚C for 10 min and left to cool down for an hour at 4˚C. Ribosomes were

crystallized by the hanging drop method at 4˚C by mixing 2-2.4 μL of ribosome solution

with 1.6 μL of well solution (95 mM Tris-Acetate pH 7.0, 95 mM KSCN, 3 mM

Mg(OAc)2, 19% glycerol, 4-4.5% w/v PEG 20,000 and 4.75 mM spermidine). Crystals

appeared within 7-10 days and reached their full size after additional two weeks.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 47: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

35

Our post-crystallization treatments included a few dehydration steps of which the

ligands were introduced at high concentrations (soaking of vacant ribosomes has been

performed for 24h and included the addition of high ligand concentrations dissolved in

the initial crystallization conditions to the resulting crystallization droplets). The

dehydration steps included the replacement of crystallization mother liqueur by a solution

containing higher concentrations of PEG 6,000 (80mM Tris-Acetate pH 7.0, 70 mM

KSCN, 10mM Mg(OAc)2, 20% v/v Glycerol, 5% w/v PEG 20,000, 6.5 mM spermidine,

7.5 mM NH4OAc, 1.4 mM Deoxy Big Chap and 2mM DTT). This solution was then

replaced step-wise, with 15 minutes breaks between steps, by solutions with increasing

concentrations of PEG 6000, reaching finally after five steps to 20% (80 mM Tris-

Acetate pH 7.0, 70 mM KSCN, 10 mM Mg(OAc)2, 18% v/v Glycerol, 5% w/v PEG

20,000, 6.5 mM spermidine, 7.5 mM NH4OAc, 20% w/v PEG 6000, without DTT,

without detergent). The final step included the introduction of dehydrated crystals for 30

min with 2 mM osmium hexamine which has been dissolved in a solution containing

20% PEG 6000. Crystals were frozen directly on the goniometer under the stream of cold

nitrogen.

Data collection was performed at the PX beamlines at the SLS, PSI facility and

crystal structure determination has been performed by using the molecular replacement

methodology with the 4.15Å solved crystal structure as a template 99

.

3.6 Leishmania Cell Culture and Promastigote Viability Assay (IC50).

The leishmanial susceptibility assay has been performed in Prof. Charles L. Jaffe's

lab at the Hebrew University in Jerusalem. In general, two strains of Leishmania were

used in the present work: L. donovani (MHOM/SD/1962/1S-Cl2d) and L. major

(MHOM/IL/2003LRC-L1025). Promastigotes were grown in complete Schneider’s

Drosophila medium containing 20% fetal calf serum and antibiotics at 26°C. Compounds

were screened for leishmanicidal activity using an alamarBlue (AbD Serotec) viability

assay essentially as previously described by Jaffe and coworkers 131

. Compounds to be

assayed were diluted in serial concentrations ranging from 0.7-400 μM (0.15-40 μM for

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 48: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

36

G418) in a complete promastigote medium, containing 1% dimethyl sulfoxide (DMSO),

and were aliquoted in triplicate (125 μL/well) into 96-well flat-bottom plates (Nunc).

Promastigotes (2.0 × 106 cells/mL; 125 μL/well) were added to each well and incubated

for 72 h at 26 °C. The alamarBlue viability indicator was added (25 μL/well) and the

plates were incubated for an additional 5 h, at which time the fluorescence (λex = 544

nm; λem = 590 nm) was measured in a microplate reader (Fluoroskan Ascent FL).

Complete medium both with and without DMSO was used as negative controls (0%

inhibition of promastigote growth). Amphotericin B (1 M, Sigma-Aldrich), a drug used

to treat visceral leishmaniasis, was included as a positive control in each plate.

3.7 The Development of a Highly Sensitive Immunoassay for 6'-OH AG Detection

3.7.1 Preparation of Polyclonal Abs

The preparation of polyclonal Abs included the conjugation of the relevant

aminoglicosidic scaffold to an immune-system-activating protein carrier, followed by

rabbits immunization and serum purification. A general scheme of immunization

conjugate preparation is described in Figure 3.1.

Figure 3.1: Synthetic scheme for the preparation of immunization\coating conjugates.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 49: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

37

3.7.1.1 Preparation of 6'-(R)-methyl-2',3-diazido-1-N-[5-oxopentanoic acid]-

paromamine (compound B, Figure 3.1).

A mixture of compound A 88

(500 mg, 1.2 mmol) and glutaric anhydride (0.29

mg, 2.5 mmol) in 20 mL anhydrous methanol was stirred under argon for 6 h at room

temperature. The reaction was monitored by TLC (EtOAc/MeOH 60:40) which indicated

completion after 6 h. The reaction mixture was evaporated under vacuum to dryness and

purified by flash chromatography, resulting in 400 mg (62% yield) of pure compound B

(Figure 3.1). The purity of the product was determined by NMR and MALDI-TOF Mass

Spectra analysis. 1HNMR (CDCl3, 500 MHz): ‘Ring I’ δH 1.26 (d, 3H, J=6.0 Hz, CH3),

3.10 (dd, 1H, J1=4.2 Hz, J2=10.5 Hz, H-2'), 3.38 (t, 1H, J=9.7 Hz, H-4'), 3.92-3.98 (m,

2H, H-5' and H3'), 4.04 (m, 1H, H-6'), 5.74 (d, 1H, J=3.1 Hz, H-1'); ‘Ring II’ δH 1.43

(ddd, 1H, J1= J2= J3=12.5 Hz, H-2ax), 2.20 (td, 1H, J1=4.5, J2=12.5 Hz, H-2eq), 3.26 (t,

1H, J=9.3 Hz, H-4), 3.52 (m, 3H, H-3, H-5, H-6), 3.78 (m, 1H, H-1). Linker peaks

appeared in the spectrum as follows: δH 1.92 (t, 2H, J=7.2 Hz), 2.29 (m, 2H), 2.36 (t, 2H,

J=7.5 Hz). 13

CNMR(CDCl3, 125.8 MHz): δC 18.0, 22.2, 33.9, 34.2, 36.2, 50.4, 61.4,

64.7, 69.3, 72.3, 74.2, 75.2, 76.2, 78.7, 80.3, 98.7 (C-1'), 175.5 (C=O), 177.1 (C=O). MS:

Calculated mass for C18H30N7O10 ([M+H]+) m/z: 504.20; measured m/z: 504.20.

3.7.1.2 Preparation of Immunization Conjugate (compound E, Figure 3.1).

A mixture of compound B (100 mg, 0.2 mmol) , N-hydroxysuccinimide (NHS)

(23 mg, 0.2 mmol) and N',N'-dicyclohexylcarbodiimide (DCC) (41.2 mg, 0.2 mmol) in

10 mL dimethylformamide (DMF) was stirred for 4h at room temperature followed by a

12 h incubation at 4˚C. The mixture was then centrifuged for 15 min at 2,500 x g at room

temperature. 1 mL of the supernatant containing 10 mg of the compound C were added

to 10 mg KLH dissolved in 4 mL of 0.13 M NaHCO3 at pH 9.2. The reaction was

incubated 12 h at 4˚C, followed by size exclusion product purification for 25 min at 3,000

x g at room temperature using Centricon 30 (Amicon, Millipore, Billerica, MA) to afford

compound D. The concentrated product D (Figure 3.1) was washed 3 times with 4 mL

of NaHCO3 solution. The final volume was adjusted to 4 mL by adding NaHCO3

solution. The reduction of azido-groups was performed by adding 100 μL

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 50: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

38

trimethylphosphine (PMe3) solution (1M solution in tetrahydrofuran - THF) and the

mixture was allowed to react 12 h at 4˚C. The final product E (referred as a compound

E, Figure 3.1) was further purified using size exclusion purification as described above,

followed by 2 washes with 5 mL NaHCO3 solution. The final volume was adjusted to 5

mL by adding double distilled water (DDW). Conjugate was stored at -20˚C pending to

use.

3.7.1.3 Immunization and Antiserum Production.

0.5 mL of immunization conjugate (compound E from the above section) were

emulsified with either Complete Freund's adjuvant (1st immunization), or incomplete

adjuvant (2nd-4th boosts) prior to immunization. The emulsion was injected

subcutaneously to two rabbits 8244 and 8245 (~1 mg conjugate were injected per rabbit

at each time point). Bleeds were collected after each boost and were tested for anti-

antigen activity using the checkerboard assay. The 3rd bleed from Rabbit 8245 has been

shown to have the highest anti-antigen activity, and was therefore used for further

experimentations.

3.7.2 The Development of a Highly Sensitive Immunoassay

The development of a highly sensitive immunoassay included the preparation of 4

coating antigens (1 similar to the immunization conjugate that was used for the

homologues assay development and 3 coating antigens for the development of 3

heterologuos assays). The coating antigens altogether with the predeveloped Abs were

used for the development of 4 indirect immunoassays. In addition, cross reactivity

experiments were designed to test the immunoassays reactivity to the various AG

derivatives used in the study and to determine the Abs affinity to various epitopes.

3.7.2.1 Preparation of NB82-OVA Coating Antigen.

NB82-OVA conjugates were prepared similarly to immunization conjugates, as

described above, except that 5 mg ovalbumin (OVA), that was dissolved in 2.5 mL

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 51: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

39

NaHCO3 solution, and then conjugated to compound B at molar ratios of 1:1, 1:2, 1:5

and 1:10. The final volume of the resulted NB82-OVA conjugate was adjusted to 1 mL

by adding 0.13 M NaHCO3, and the pH of the solution was adjusted to 9.6 by adding

dilute solution (0.01 M) of NaOH. Protein content has been determined to be 5 mg mL-1

using the Bradford assay (BioRad protein assay, BioRad, Hercules, CA) and the resulting

conjugates antigen activity was evaluated towards 3rd bleed antiserum from Rabbits 8244

and 8245 using the checkerboard assay. The 1:1 NB82-OVA conjugate crossed with 3rd

bleed antiserum derived from Rabbit 8245 has been shown to have the most sensitive

activity, and was therefore used for further experimentation. The conjugates were stored

in aliquots at -20˚C.

3.7.2.2 Preparation of NB82/84/124-GTA-OVA Coating Antigens

To prepare coating antigens for heterologous-cross reactivity assays we used

glutaraldehyde (GTA) coupling method as described by Chen at al. 132

. In brief, 20 mg

OVA was dissolved in 10 mL of 0.01 M phosphate-buffered saline (PBS) at pH 6.5 (the

pH adjustment was performed prior to the addition of OVA by adding dilute solution,

0.01 M, of HCl). 20 mg of each compound (NB82/84/124 sulfate) was added to the

solutions and stirred for a few min at r.t. 1.5 mL of freshly prepared 1% glutaraldehyde

solution was added in a drop-wise manner using a 1 mL syringe equipped with a 0.5 mm

x 16 mm needle. The reaction mixture was allowed to be stirred at r.t for 15 min, of

which yellow color indicated the imine formation. Reduction of imines to amines has

been performed by adding 115 mg sodium borohydride, followed by 12 h incubation at

4˚C. The final product has been purified using Centricon 30 for 30 min at 3,000x g,

followed by 2 washes with 1 mL 0.01 M PBS pH 7.2. The final volume of conjugates has

been adjusted to 1 mL using PBS pH 7.2, and their protein content has been determined

to be 17.5-19 mg mL-1

using the Bradford methodology. Conjugate antigen reactivity was

tested against 3rd bleed antiserum from Rabbits 8244 and 8245 using the checkerboard

assay. The conjugates were stored in aliquots at -20˚C pending to use.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 52: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

40

3.7.2.3 NB82 Indirect Competitive ELISA

The assay developed was an indirect competitive ELISA in which tested samples

or standard solutions were allowed to compete with an immobilized coating antigen for

antiserum binding. The assay has developed by using microtiter 96 well plates (F96

Maxisorp, Nunc Immuno Plate, Roskilde, Denmark). Wells were coated with 100 μL

NB82-OVA (1:1) conjugate, diluted 1:8,000 (62.5 ng conjugate per well) in 0.5 M

carbonate buffer, pH 9.6 (CB). After an overnight incubation at 4˚C, wells were washed 3

times with PBST (defined as PBS pH 7.2 containing 0.3% [v/v] Tween-20). 50 μL of

unknown samples or standard solutions (in PBS, mice serum or Fetal bovine serum -

FBS, Biological Industries, Israel) were added to the wells, followed by the addition of

50 μL of antiserum (Rabbit 8245) diluted 1:80,000 in PBST. The standard samples were

comprised of 12 serial dilutions of NB82, ranging from 10-0.0049 μg mL-1

dissolved in

PBS or serum. Plates were incubated overnight at 4˚C, and washed 3 times with PBST.

100 μL secondary antibody (2nd Ab) conjugated to horseradish peroxidase (HRP) (anti-

rabbit HRP conjugated, Sigma), diluted 1:20,000 in PBST were added to each well.

Plates were incubated for 2 h at r.t, rinsed with PBST, and tested for HRP activity by the

addition of 100 μL substrate solution - 3,3',5,5'-tetramethyl benzidine (TMB substrate

chromagen, Dako, Glostrup, Denmark). The reaction was stopped after 20 min by adding

50 μL of 4 N sulfuric acid, and the absorbance was measured with ELISA reader (Spectra

Max M2, Molecular devices, Sunnyvale, CA) at 450 nm. In all experiments a 5 mg mL-1

OVA solution in PBS has been used as a negative control to evaluate and normalize the

non-specific binding of antisera. Non-specific binding values obtained were almost

negligible in all experiments. Wells coated with coating antigen that were not introduced

with a free antigen sample were used as a positive control and determined the 100%

binding of antisera.

3.7.2.4 Cross Reactivity Experiments

Cross reactivity values of antisera with various AG were determined using the

heterologous indirect ELISA as described above by adding the subjected compounds

(instead of NB82) at 24 serial dilutions ranging from 10 mg mL-1

to 1.19 ng mL-1

, except

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 53: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

41

for NB84, of which serial dilution ranged from 1 mg mL-1

to 0.119 ng mL-1

. All AG

tested were in their sulfated form and were dissolved and diluted in PBS or serum. The

standard solutions of the compounds NB82, NB84 and NB124 were shown to be stable in

PBS and standard serum (FBS) solutions that were frozen (-20˚C) for a few months and

demonstrated similar values using at least 5 repeated freezing/thawing cycles.

3.7.2.5 NB82/84/124 Indirect Competitive ELISA

In order to enhance antiserum sensitivity towards hetero-compounds that share

similar epitopes with the derivate of which antiserum antibodies has been produced

against, an indirect heterologous-assay has been developed. The assay has been

performed similarly to the indirect assay for NB82 as described in section 2.8, except for

the coating antigens of which for the NB82 heterologous-test, NB82-GTA-OVA

conjugate diluted 1:128,000 in CB was used as the coating antigen; for the NB84

heterologous-test, NB84-GTA-OVA conjugate diluted 1:64,000 in CB was used as the

coating antigen; and for the NB124 heterologous-test, NB124-GTA-OVA conjugate

diluted 1:16,000 in CB was used as the coating antigen. Solutions that contained 17.5-19

mg mL-1

OVA in PBS were used for coating negative control wells. Cross reactivity

experiments have been performed as indicated in section 3.7.2.4.

3.7.2.6 Data Analysis

The sensitivity (I50) and limit of detection (I20) values were obtained from fitting

concentration vs. % binding curves to a log-logit plot using the Origin software, version

8.5 (Mirocal Software, North Hampton, MA, USA).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 54: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

42

3.7.3 Determination of NB Compounds Levels in Mice Serum

Determination of NB compounds serum levels included an intraperitoneal (IP)

injection of NB84/NB124 to mice and collection of blood samples at various time points.

The protocol included the IP injection of 28.5 μg/gr body weight of the relevant NB

compound to 7 male mice (C57/BL, 2.5-3.5 months old), followed by collection of fresh

blood samples (~200 μL each) at 7 time points (5, 10, 20, 30, 40, 50, 60 min). Blood was

collected to sterile tubes containing 20 μL of 50 mM EDTA pH 8.0. Serum was further

extracted by centrifugation at 2,000x g at 4˚C for 10 min. NB compounds content in

serum samples was determined by using the pre-developed microtiter assay, as described

above, when compared to a linearized calibration curve transformed to a log-logit plot

using the Origin software, version 8.5. Each sample was tested in duplicates at five

dilutions. Serum recovery ratios were determined by spiking experiments of which whole

blood samples derived from untreated mice were spiked with known amounts of the

relevant aminoglycosidic derivatives. Blood samples were incubated for 1 h at 4˚C

followed by serum preparation by centrifugation as described above.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 55: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

43

4. Results and Discussion

4.1 Deciphering Aminoglycosides Mechanisms of Action in Eukaryotes

To explore the interactions of AG within their binding site in eukaryotes we used

complexes of various AG representatives with double stranded rRNA constructs

mimicking their putative binding site. The rRNA constructs contained one or two binding

sites corresponding to the sequences of the eukaryotic A-sites (Figure 4.1) and were

similar to previously reported constructs used for the exploration of AG interactions in

bacterial and human systems by crystallographic means 104, 113, 118

.

Figure 4.1 The rRNA constructs used to explore AG interactions: A. Human double A-site; B. Protozoa

double A-site; C. Human single A-site. Sequences corresponding to the natural A-site are highlighted in

dashed red squares. A/G1408 - orange, A1491 - green, A1492 - blue and A1493 - red. The rRNA residues

are numbered according to the numbering used in E. coli 16S rRNA.

Human

Double A-site model

Protozoa

Double A-site model

Human

Single A-site model

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 56: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

44

The crystallographic studies included RNA purification, complex generation,

crystallization and structure determination and are fully described in the materials and

methods section.

4.1.1 rRNA Purification, Complex Generation and Crystallization

The RNA oligomers used in this study were chemically synthesized. Preparation

of active RNA-AG complexes included: RNA deprotection, anion exchange purification,

SPE desalting, RNA denturation/annealing cycle and finally complexation with AG.

These steps are fully described in the materials and methods section and a representative

example of the RNA purification steps is illustrated in Figure 4.2.

Figure 4.2. RNA deprotection and purification. A. Deprotection scheme. B. FPLC diagram. C. PAGE

analysis of FPLC RNA containing RNAs. Lanes highlighted in red are the ones used for further desalting

and complexation procedures.

Our initial crystallization trials were performed using a complex of the human

double A-site model (Figure 4.1A) with one of the lead read-through inducing

compounds developed in our lab, NB54 (Figure 1.7). An initial crystallization screen

was designed to test various mono-valent and di-valent ions as well as 2 different

polyamines and precipitating agents known to mediate RNA crystal growth Table I

(Appendix). Screening was done by an automatic robot screen using a 96 well sitting

drop crystallization plate, in the presence of 2 mM RNA and 4 mM AG solutions at pH

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 57: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

45

7.0. Crystals appeared after 3-4 days in 9 out of 96 conditions (Figure 4.3). The best

crystals were obtained in the presence of 250 mM LiCl and 100-250 mM KCl over a 75

μLreservoir containing 40% MPD or 20% PEG3350 (Table I, Appendix). Additional

optimization steps were designed to test a wider range of monovalent salt concentrations

as well as combinations of high concentrations of mono-valent salts with lower

concentrations of di-valent ions (Mg+2

) (Table II, Appendix). The optimization screen

was performed in a wide pH range used to test pH influence on crystal growth

(experiments were performed in the range of pH 5.0-7.0). The optimization steps were

performed in 24 well plates using the hanging drop crystallization method, and resulted in

the emergence of large crystals (100-250 μm) which were highly suitable to X-ray

analysis (Figure 4.4). Our optimization results indicated that crystals grown from the

KCl conditions had better morphologies than the ones that emerged in the presence LiCl

or NH4Cl conditions. In addition MPD was found to be a better precipitant than PEG.

Figure 4.3. Crystals emerged from an initial screen of eukaryotic rRNA duplex in complex with NB54.

The crystallization conditions were A. 250 mM LiCl over a 20% PEG3350 reservoir (condition A6); B. 100

mM KCl over a 40% MPD reservoir (condition C2); C. 250 mM KCl over a 20% PEG3350 reservoir

(condition C6); D. 250 mM KCl over a 40% MPD reservoir (condition C9); E. 100 mM KCl over a 20%

PEG3350 reservoir (condition C11); F. 250 mM NH4Cl over a 20% PEG3350 reservoir (condition D12).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 58: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

46

Figure 4.4. Crystals obtained from an optimization screen of eukaryotic rRNA duplex in complex with

NB54. A. 50 mM KCl. B. 100 mM KCl. C. 150 mM KCl. D. 200 mM KCl. E. Frozen crystal of 200 mM

KCl on a nylon cryo-loop. F. Diffraction pattern of the frozen crystal as obtained in ID14-1 ESRF.

4.1.2 Crystal Structure Determination of Human A-site rRNA Complexes with AG

4.1.2.1 The Double A-site Model

An X-ray analysis of dozens of crystals indicated that best diffraction was

obtained from crystals containing higher salt concentrations (150-200 mM KCl). These

crystals diffracted up to 1.96 Å in the presence or absence of 40% MPD as a cryo-

protecting solution. Crystal data of 3 representatives (CYT1, CYT2 and CYT3) is

summarized in Table 4.1. The resulting crystal structures indicated two distinct

conformations of the two A-sites present in the rRNA model (Figure 4.5). One such

conformation was earlier described as an ‘ON’ state 121

(upper binding site, Figure 4.5 A-

C) where the two conserved adenine residues, A1492 and A1493, are flipped out from

the A-site helix; The conformation observed within the second binding site was denoted

as an ‘OFF’ state, where A1493 and A1491 are flipped out and A1492 is directed towards

the helical groove (Figure 4.5 A,B,D). The two observed conformations are believed to

represent the two possible conformational states in the ribosomes decoding site mediating

translation continuation and arrest in the presence of cognate or near/non-cognate tRNA

molecules, respectively.

A. B. C.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 59: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

47

Table 4.1. Crystal data, data collection and data refinement statistics – NB54

NB54-cyt 3 NB54-cyt 2 NB54-cyt 1

150 mM KCl 200 mM KCl 150 mM KCl Crystallization Conditions

- 40% MPD 40% MPD Cryo-Protectant

Crystal Data P21212 P21212 P21212 Space group

a=45.36

b=47.16

c=56.62

α=90°

β=90°

γ=90°

a=45.7

b=47.1

c=55.6

α=90°

β=90°

γ=90°

a=40.93

b=45.37

c=54.27

α=90°

β=90°

γ=90°

Unit cell parameters (Å/°)

1 1 1 Za

Data Collection

ID14-1 ESRF

Grenoble, France

ID14-1 ESRF

Grenoble, France

Rigaku Raxis IV++

Tel Aviv university

Beamline

0.934 0.934 1.541 Wavelength (Å)

35-2.5 36-1.96 30-2.7 Resolution (Å)

28,162 58,386 11,080 Observed reflections

4,341 8,852 3,033 Unique reflections

99.9 99.9 99.9 Completeness (%)

100 100 100 in outer shell (%)

5.7 7.5 5.4 Rmergeb (%)

13.5 35.5 38 in outer shell (%)

20.1 13.3 14.2 Mean ((I)/sd(I))

10.5 4.7 2.5 in outer shell (%)

Structure Refinement

35-2.5 36-1.96 30-2.7 Resolution range (Å)

4,124 8,831 3,021 Reflections

22.5/26.3 21.2/26.4 21.9/22.9 R-factorc / Rfree

d (%)

920 920 912 Number of RNA atoms

- - 2 Number of ligand molecules

0 (*) 113 3 Number of water molecules

R.M.S.D

0.014 0.005 0.01 Bond length (Å)

2.65 0.896 2.25 Bond angles (°)

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the observed and calculated structure factor

amplitudes, respectively. d Calculated using a random set containing 5% of observations that were not included in refinement.

* The structure was not fully refined due to the lack of ligand in the electron density.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 60: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

48

Figure 4.5. Structures of rRNA crystals that were crystallized in the presence of a potent read-through

inducer – NB54. A. A 2D representation of the human cytoplasmic double A-site constructs as obtained in

the X-ray crystal structures. B. The double A-site construct 3D structure demonstrating two remote

conformational states of the putative binding sites: an ‘ON’ (upper site) vs. an ‘OFF’ (lower site) state

conformation. C. A closer look into the ‘ON’ state conformation with the two conserved adenine residues

(A1492 - blue and A1493 - red) budging out from the helical core. D. The ‘OFF’ state conformation with

A1491 (green) and A1493 (red) flipping out and A1492 (blue) directed towards the inner part of the helical

core. E. A 3D representation of CYT1 structure with some possible binding sites of NB54. F. Binding of

NB54 2 rings to the ‘ON’ state, Fo-Fc electron density maps are contoured at 2.0σ around the AG. G.

Binding of NB54 to the models GC pairs region, Fo-Fc electron density maps are contoured at 2.0σ around

the AG. H. Superposition of rRNA residues of CYT1 (green), CYT2 (blue), CYT3 (yellow) and an empty

rRNA duplex (PDB ID 2FQN – red). R.M.S.D values for CYT1, CYT2, CYT3 and 2FQN are 0.90Å,

0.40Å and 0.34Å, respectively.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 61: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

49

Unfortunately, an indication of the ligand’s presence was observed in only one of

the three structures solved (CYT1). The extra electron density obtained in this structure

indicated the presence of two NB54 molecules in the double A-site construct. With one

ligand molecule bound to the deep/major groove of the RNA duplex stabilizing the 'ON'

state conformation, and the second bound between the G=C pairs separating the two A-

sites (Figure 4.5 E-G). However, the extra electron densities for both ligands exhibited

some rather low sigma values (σ=2.0 in Fo-Fc maps) indicating low occupancy of the

ligand molecules within the crystal structure, and making it almost impossible to

determine the exact location of the 3 sugar rings within the structure. In addition,

superposition of the three solved crystal structures with a similar rRNA model

crystallized in the absence of an AG molecule 121

indicated that all four structures share a

great conformational and packing similarity (Figure 4.5 H); Implying a rather low

occupancy of ligands’ actual presence in the CYT1 structure.

The AG:A-site stoichiometric ratio used in our experiments was 1:1. Similar

ratios have previously been used for the complex generation of various AG with bacterial

A-sites. However, taking under considerations that AG affinity towards human ribosomes

is 3 orders of magnitude lower than in bacteria 133

we suggested that the low occupancy

values obtained in our structures were as a result of low ligand affinity towards the

human A-site. To further explore this notion, we generated rRNA-NB54 complexes at

higher AG:A-site ratios. Crystal obtained up to 10:1 ratios and were found suitable for X-

ray analysis (higher ratios resulted in the immediate appearance of a thick precipitate in

all tested conditions). In addition to the increase in ligand’s concentration we added the

ligand at as high a concentration as possible to the cryo solution to make sure that the

ligand does not leach out from the crystal during cryo-protection steps. Four examples of

crystal data are presented in Table 4.2. As can be seen, all crystals shared similar

symmetry and unit cell parameters. Unfortunately, the increase in ligands concentration

did not result in any ligand’s indication at the obtained electron density maps and the

resulting structures were identical to the ones reported with lower ligand concentrations.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 62: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

50

Table 4.2. Crystal data, data collection and data refinement statistics

NB54-cyt 7 NB54-cyt 6 NB54-cyt 5 NB54-cyt 4

A-site-NB54 1:10 A-site-NB54 1:8 A-site-NB54 1:4 A-site-NB54 1:2 Crystallization Conditions

40% MPD + 8mM

NB54 soak (2 h)

40% MPD + 8mM

NB54 soak (2 h)

40% MPD + 8mM

NB54 soak (2 h)

40% MPD + 8mM

NB54 soak (2 h) Cryo-Protectant

Crystal Data P21212 P21212 P21212 P21212 Space group

a=45.36

b=47.16

c=56.62

α=90°

β=90°

γ=90°

a=40.93

b=45.37

c=54.27

α=90°

β=90°

γ=90°

a=45.7

b=47.1

c=55.6

α=90°

β=90°

γ=90°

a=45.8

b=47.44

c=57.75

α=90°

β=90°

γ=90°

Unit cell parameters (Å/°)

1 1 1 1 Za

Data Collection

ID23-2 ESRF

Grenoble, France

ID14-4 ESRF

Grenoble, France

ID23-2 ESRF

Grenoble, France

ID14-4 ESRF

Grenoble, France

Beamline

0.873 0.9394 0.873 0.9394 Wavelength (Å)

36-2.5 56.7-2.8 47-2.8 57.75-3.0 Resolution (Å)

13,030 8,588 16,018 12,002 Observed reflections

4,504 2,874 3,375 2,592 Unique reflections

92.7 91.2 99.4 99.4 Completeness (%)

93.6 93.7 99.9 99.6 in outer shell (%)

7.9 8.9 10 8.8 Rmergeb (%)

12.8 11.3 59.4 12.5 in outer shell (%)

9.7 10 10.5 10.2 Mean ((I)/sd(I))

3.9 4.8 3.4 5.7 in outer shell (%)

Structure Refinement

36-2.5 56.7-2.8 57.6-2.8 57.75-3.0 Resolution range (Å)

4,206 2,719 3,214 2,465 Reflections

25.6/26.1 25.1/27 24.1/26 20/22 R-factorc / Rfree

d (%)

920 920 920 920 Number of RNA atoms

1 - - 1 Number of ligand molecules

1 - - - Number of water molecules

R.M.S.D

0.011 0.012 0.012 0.013 Bond length (Å)

2.27 2.22 2.43 2.452 Bond angles (°)

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the observed and calculated structure factor

amplitudes, respectively. d Calculated using a random set containing 5% of observations that were not included in refinement.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 63: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

51

In parallel to NB54 we crystallized complexes of two additional synthetic

derivatives, NB74 and NB124 (Figure 1.7). These derivatives were synthesized in our

lab and were marked as highly potent read-through inducers 88, 91

. Our crystallization

trials resulted in dozens of crystals suitable for X-ray analysis. Crystals morphologies

were similar to the one present in Figure 4.4 C, as well as the unit cell parameters and

crystal’s symmetry (Table 4.5). Out of 10 solved structures of NB74-A-site complexes in

various rRNA:AG ratios, an indication of ligand's presence appeared only in 2 crystals:

one crystal with two ligands bound (one at the deep/major groove of the RNA duplex

stabilizing the 'ON' state conformation, and the other bound between the G-C pairs

separating the two A-sites, as can be seen in Figure 4.6); and a second crystal with only a

single ligand bound in the G-C pairs region. The ligand's electron density was stronger

than the one obtained for NB54; therefore presenting a higher probability to the ligands

presence. 2Fo-Fc maps (σ=1.0) for the NB74-cyt2 complex, potentially containing two

bound ligands, are presented in Figure 4.6 C,D. As can be seen electron density was

much stronger for the ligand bound at the G-C pairs vicinity, which does not consist a

part of the natural A-site sequence; therefore defined as non-specific binding. Such non-

specific interactions with the non-natural constructed G-C pair region were previously

reported in the studies of both natural and semi-synthetic AG derivatives in complex with

human and bacterial A-site constructs 92, 110, 112

. The electron density for the ‘ON’ state

ligand was much lower than the one present for the G-C pairs bound ligand, and

encountered similar problems as previously explained for the NB54 structure (Figure

4.7). For the NB124 complexes, no ligands were observed in the structures’ electron

density. Samples of crystals data are presented in Table 4.3.

With only a vague indication for ligand’s presence we wondered if due to the

minimal binding site available in our model system the binding site conformation as well

as the ligand’s binding pattern is affected by crystal packing interactions. An in depth

analysis of the crystal packing indeed indicated a stabilization of the obtained adenine

conformations by - stacking interactions with adenine residues bulging out from

adjacent molecules located within the crystal Figure 4.8. These indications suggest that

the two distinct conformations observed result from crystal packing interactions and are

not mediated by the ligand's presence.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 64: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

52

Table 4.3. Crystal data, data collection and data refinement statistics

NB124-cyt 1 NB74-cyt 2 NB74-cyt 1

A-site-NB124 1:1 A-site-NB74 1:4 A-site-NB74 1:1 Crystallization Conditions

40% MPD 40% MPD +

NB74 soak (2 h)

40% MPD Cryo-Protectant

Crystal Data P21212 P21212 P21212 Space group

a=47.36

b=47.63

c=56.86

α=90°

β=90°

γ=90°

a=47.66

b=47.68

c=57.5

α=90°

β=90°

γ=90°

a=48.03

b=48.77

c=58.13

α=90°

β=90°

γ=90°

Unit cell parameters (Å/°)

1 1 1 Za

Data Collection

NarµX X-ray System

Ben-Gurion university

ID14-4 ESRF

Grenoble, France

ID23-2 ESRF

Grenoble, France

Beamline

1.541 0.9394 0.873 Wavelength (Å)

56.8-3.0 57.5-2.77 37.03-2.8 Resolution (Å)

6,778 10,742 6,953 Observed reflections

2,540 4,989 3,248 Unique reflections

92.2 94.8 83.0 Completeness (%)

94.3 96.8 86.3 in outer shell (%)

11.3 7.5 12.9 Rmergeb (%)

27.5 43.4 29.2 in outer shell (%)

6.1 7.3 5.5 Mean ((I)/sd(I))

2.8 2.5 2.7 in outer shell (%)

Structure Refinement

56.8-3.0 57.5-2.77 37.035-2.7 Resolution range (Å)

2,406 4,206 3,098 Reflections

21/24.8 24.9/27.5 22.1/23.06 R-factorc / Rfree

d (%)

920 920 920 Number of RNA atoms

- 2 1 Number of ligand molecules

- 12 - Number of water molecules

R.M.S.D

0.013 0.012 0.011 Bond length (Å)

2.58 2.34 2.24 Bond angles (°)

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the observed and calculated structure factor

amplitudes, respectively. d Calculated using a random set containing 5% of observations that were not included in refinement.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 65: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

53

Figure 4.6. NB74-human cytoplasmic A-site complex. A. A 2D representation of the human cytoplasmic

double A-site constructs as obtained in the X-ray crystal structure in the presence of NB74. Putative

binding sites for the two ligands are marked as Lig1, Lig2, respectively. Each A-site occupies a different

conformation: An ‘ON’ (upper) vs. an ‘OFF’ state (lower). B. The double A-site construct with two NB74

molecules (yellow) bound: one ligand is bound within the upper binding site demonstrating an ‘ON’ state

conformation; while the other ligand (lower) is bound to the G-C pairs region. A1491 is highlighted in

green, A1492 in blue and A1493 – red. C. 2Fo-Fc electron density maps (contoured at 1.0σ around the AG)

of lig1. D. 2Fo-Fc electron density maps (contoured at 1.0σ around the AG) of lig2. A1491 is highlighted in

green, A1492 in blue and A1493 – red.

Figure 4.7. B-factor analysis of NB54-

Cyt1(A) and NB74-Cyt2 (B). Residues

colored in red/orange/yellow indicate high

B-factor values; thus implying for low

electron density or high residual flexibility.

Residues colored in blue/green indicate low

B-factor values; implying for a rather high

electron density or low flexibility.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 66: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

54

Figure 4.8. Crystal packing. A. Stabilization of a flipped out A1491 (green) by A1492 (blue) belonging to

an adjacent rRNA molecule in the crystal packing by - stacking interactions. Stacking interactions are

marked in black arrow (distance = 3.9Å). B. Stabilization of a flipped out A1491 (green) by A1493 (red)

of an adjacent molecule in the crystal packing by - stacking interactions.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 67: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

55

Previous studies exploring the interactions of strong inhibitors of human

ribosomes, exhibiting no read-through activity 92

, used a similar double A-site construct

and resulted in a different crystal packing with a strong indication for ligand’s presence.

As a positive control to our experimentations we chose Apramycin, an efficient anti-

bacterial agent that is known to bind the A-site but exhibits no read-through activity in

eukaryotes 92

. Generation of a 1:1 ratio complex of Apramycin with the human double A-

site model resulted in the emergence of diamond shaped crystals (Figure 4.9 A) that

diffracted up to 2.9Å, and exhibited crystal symmetry and unit cell parameters which are

different than the ones present for the rRNA read-through inducing derivatives

complexes (Table 4.4). As can be seen in Figure 4.9, the two model A-sites occupied the

same conformational state, the ‘OFF’ state, with two Apramycin molecules bound, one to

each A-site and two cobalt-hexammine (Co-Hex) molecules bound at the G-C pairs

region. Similar Co-hex interactions were previously found in various crystal structures of

human and bacterial models crystallized in the presence of Co-Hex 19, 118, 121

.

Figure 4.9. A. Crystals Apramycin-eukaryotic-rRNA duplex. B. A 2D representation of the human

cytoplasmic double A-site constructs as obtained in the X-ray crystal structure with both A-site at the

‘OFF’ state conformation. C. The double A-site construct 3D structure with two Apramycin molecules

(yellow) bound, one ligand molecule to each A-site. Both A-sites occupy the ‘OFF’ state conformation

where A1491 (green) and A1493 (red) are flipped out and A1492 (blue) is directed towards the helical

core. Cobalt hexamine molecules are highlighted in pink. D. Apramycin bound to a single ‘OFF’ state.

2Fo-Fc electron density maps are contoured at 1.0σ around the AG.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 68: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

56

Table 4.4. Crystal data, data collection and data refinement statistics for the Apra-cyt crystal

Apra-cyt

Crystal Data P31 Space group

a=32.56; b=32.56; c=111.09 ; α=90°; β=90° γ=120° Unit cell parameters (Å/°)

1 Za

Data Collection

ID29 ESRF

Grenoble, France

Beamline

0.9763 Wavelength (Å)

37.0-2.9 Resolution (Å)

5,988 Observed reflections

2,056 Unique reflections

93.9 Completeness (%)

95.4 in outer shell (%)

17.2 Rmergeb (%)

51.1 in outer shell (%)

4.7 Mean ((I)/sd(I))

2.6 in outer shell (%)

Structure Refinement

37.0-2.9 Resolution range (Å)

1,945 Reflections

24.0/26.0 R-factorc / Rfree

d (%)

940 Number of RNA atoms

2 Number of ligand molecules

- Number of water molecules

R.M.S.D

0.021 Bond length (Å)

3.0 Bond angles (°)

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the

observed and calculated structure factor amplitudes, respectively.

d Calculated using a random set containing 5% of observations

that were not included in refinement.

Figure 4.10. Crystal packing of

the Apra-cyt complex, showing a

stabilization of a flipped out

A1491 (green) by A1493 (green)

belonging to an adjacent rRNA

molecule in the crystal packing by

- stacking interactions.

Stacking interactions are marked

in black arrow (distance = 4Å).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 69: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

57

The electron densities for both Apramycin molecules bound were well ordered

and indicated the binding of the two ligand molecules at similar conformations in both A-

sites (Figure 4.9). The Fo-Fc maps prior to ligand's introduction indicated clearly the

ligand’s presence and could easily be used for the determination of ligand’s configuration

within the crystal structure. Crystal packing analysis of the resulting structure

demonstrated the stabilization of the flipped out adenine residues, A1491 and A1493, by

the same residues bulging out from adjacent rRNA molecules; indicating that these

residues might be more flexible and occupy additional conformations when in solution.

In-vitro experiments previously performed in our group indicated that Apramycin

as well as several read-through inducers such as Paromomycin and various NB

compounds are similarly capable of inhibiting eukaryotic ribosomes at the low micro-

molar range 92

. Similar experiments performed in prokaryotes demonstrated that AG such

as Paromomycin and Apramycin have a 3 order of magnitude higher capability to inhibit

prokaryotic ribosomes when compared to eukaryotes 92

. Combining this knowledge with

our indefinite indication for the presence of the 3 tested NB compounds within the crystal

structure, altogether with packing interference and the positive control demonstrating a

strong binding of Apramycin to the models A-sites we assumed that in order to get a

definite indication for read-through inducers binding to the A-site we will need to either

use extremely different crystallization conditions that will induce a different crystal

packing or try and search for a new model.

Our search for new crystallization conditions included the use of commercial

screening kits for RNA crystallization such as Natrix and Nucleic Acid Mini Screen by

Hampton research. Unfortunately, no new crystal forms emerged from the crystal screen,

and crystals emerged only in conditions which were similar to the ones that resulted in

crystal growth in our previous crystallization experiments. A few crystallization

experiments performed in conditions using extreme pH values (pH 5) (Table II,

appendix) resulted in the emergence of two different crystal morphologies at the same

crystallization droplet (Figure 4.11 A). The crystals emerged from complexation of the

natural read-through inducer, G418 (Figure 1.1), with the human double A-site construct

and appearance of crystals with the new morphology was after two month of incubation

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 70: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

58

at 20°C. X-ray analysis of both crystal types revealed that indeed crystals that

demonstrated the new morphology had different unit cell parameters than the previously

obtained ones (Table 4.5); whereas crystals that demonstrated a similar morphology to

the previously reported ones emerged after 4 days and were found to be identical to the

previous one and ligand free. The structure of the new crystal form could be easily

determined by using molecular replacement with bacterial A-site models complexed with

G418, and both the Fo-Fc maps prior to ligand’s introduction and 2Fo-Fc maps after its

addition clearly indicated the ligand’s binding to one of the human A-sites (Figure 4.11).

However, the relatively small crystal size (~20 μm) resulted in a fast deterioration of

tested crystals upon X-ray exposure; therefore resulting in the need to merge a large

number of data sets for data interpretation (data from 5 crystals were merged to build a

single structure). The resulting resolution was poor (up to 3.2Å) and the electron density

for the unpaired adenine residues (A1491-3) at the additional unoccupied binding site

was disordered; therefore making it impossible to determine their orientation and

finalizing the crystal structure’s determination.

Figure 4.11. G418-human

cytoplasmic A-site complex at

pH 5.0. A. Crystals emerged

from crystallization experiments

at pH 5.0. Two crystal

morphologies were obtained in

a single crystallization droplet.

The round crystals highlighted

in red are those with the new

symmetry-unit cells parameters.

The orthorhombic crystals at the

right lower corner are identical

in symmetry and unit cell

parameters to the previously

obtained crystals in pH 7.0.

B. The occupied A-site in the absence of G418. 2Fo-Fc electron density maps are contoured at 1.0σ - blue; Fo-Fc

electron density maps are contoured at 3.0σ – red (maps are presented in COOT). C. The occupied A-site after the

addition of G418. 2Fo-Fc electron density maps are contoured at 1.0σ - blue; Fo-Fc electron density maps are

contoured at 3.0σ – red (maps are presented as calculated in COOT). D. The occupied A-site 3D structure. G418 is

highlighted in yellow, A1491-green, A1492 – blue and A1493 – red.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 71: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

59

Table 4.5. Crystal data, data collection and data refinement statistics

G418-Cyt – pH 5.0

G418-cyt

Na cacodylate pH 5.0 Crystallization Conditions

40% MPD Cryo-Protectant

Crystal Data P21212 Space group

a=30.49

b=85.86

c=45.46

α=90°

β=90°

γ=90°

Unit cell parameters (Å/°)

1 Za

Data Collection

ID23-1 ESRF

Grenoble, France

Beamline

0.9763 Wavelength (Å)

50.0-3.2 Resolution (Å)

5,988 Observed reflections

2,056 Unique reflections

93.9 Completeness (%)

95.4 in outer shell (%)

17.2 Rmergeb (%)

51.1 in outer shell (%)

4.5 Mean ((I)/sd(I))

2.5 in outer shell (%)

Structure Refinement

50.0-3.2 Resolution range (Å)

1,772 Reflections

33/38 R-factorc / Rfree

d (%)

898 Number of RNA atoms

1 Number of ligand molecules

- Number of water molecules

R.M.S.D

0.011 Bond length (Å)

1.797 Bond angles (°)

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the observed and calculated structure factor

amplitudes, respectively. d Calculated using a random set containing 5% of observations that were not included in refinement.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 72: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

60

Unfortunately, despite our efforts to improve crystal quality we did not obtain

better diffracting crystals. Our failure to improve the resulting crystals could mainly be

attributed to the long crystallization period (at least two months) and the very low

reproducibility of the crystallization method. The optimization trials included the addition

of various additives such as glycerol and PEGs to the crystallization solutions, as well as

the use of a wide range of MPD concentrations at both reservoir and crystallization

solutions. In addition both macro- and micro- seeding experiments were performed (these

trials are fully described in the materials and methods section).

However, using such extreme pH values (5 and below) in RNA crystallization

is usually not recommended due to adenine protonation. Such protonation events might

induce some un-natural base-paring interactions that are rarely possible under

physiological conditions; therefore inducing an experimental artifact (Figure 4.12 D). In

addition, low pH values also induce protonation of additional amino groups in the ligand,

which might also enhance some non-natural interactions with the rRNA construct.

Despite the G418 data incompleteness, the bound A-site electron density is very clear and

therefore we could use it to further explore the resulting structure. Our explorations

indeed indicated the presence of possible interactions of a protonated adenine residue,

A1491, with a cytosine residue located at the opposite rRNA strand (C1409). Such

interactions resulted in the generation of a pseudo-canonical base-pair between two

nucleotides that usually don’t interact in canonical modes, making the A-site geometry

much more similar to the one present in bacterial A-sites (r.m.s.d 0.4 Å; Figure 4.12 A).

Superposition of the C1409:A1491 base pairs obtained in the ‘ON’ state A-sites of the

two structures (pH 5.0 and 7.0) indeed indicate a large difference in base paring

interactions (Figure 4.12 C). As a matter of fact, although the two bound A-sites occupy

the ‘ON’ state conformation where the two conserved adenine residues are bulged out,

their actual geometry is vastly different (r.m.s.d 2.604 Å; Figure 4.12 B). These

geometrical differences might be attributed either by the intermolecular interactions

inside the crystal packing or by the absence of solid base-pair interactions of the

C1409:A1491 pair in the pH 7.0 structure. The significant geometrical similarity and

ligand’s orientation of the pH 5.0 structure with the bacterial binding site, altogether with

its strong indication for ligand’s presence, implicates the importance of such geometry

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 73: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

61

for the ligands binding and might explain why the pH 7.0 structure presented only a

vague indication to ligand’s presence, if any.

Figure 4.12. Analysis of the pH 5.0 ‘ON’ state structure. A. Superposition of bacterial A-site (blue) and

human A-site at pH 5.0 (orange) with a bound G418 molecule. B. Superposition of pH 7.0 (green) and pH

5.0 (orange) ‘ON’ states. C. Superposition of the conformation of the C1409:A1491 pair of the pH 5.0

structure (orange) indicating a pseudo Watson-Crick pair, and the pH 7.0 (green) indicating a shifted

conformation. D. Pseudo Watson-Crick pair possible between adenine and cytosine at low pHs.

C1409 A1491

A1492

A1493

A1493

A1493

A1492

A1492

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 74: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

62

To further explore the importance of binding site geometry to ligand’s binding

and avoid working in extreme pH conditions and long crystallization periods, we decided

to generate a mutant A-site model containing a single mutation at position 1409, C1409U

(Figure 4.1 B). We hypothesized the such a mutation would induce the generation of a

‘real’ Watson-Crick pair that is not pH dependent, and will result in a geometry that is

similar to the one obtained in pH 5.0 without inducing a major difference in the binding

site sequence or the ligand’s charge. Interestingly, such A-site sequence is identical to the

w.t. A-site of several parasites, such as Leishmania, which are known to induce severe

disease symptoms in mammals. The obtained results will be described in chapter 4.1.4.

4.1.2.2 The Single A-site Model

Additional crystallization trials of human A-site in complex with AG included the

use of an extended single A-site model (Figure 4.1 C). The model is composed of two

rRNA strands with sequences that correspond to an extended sequence of the human A-

site with some additional nucleo-base-pairs used to stabilize the double stranded model.

The addition of the 3’-cytosine and guanine residues was intended to induce “sticky”

ends in order to facilitate crystal growth. A similar model was previously used to explore

the interactions of Apramycin with the human A-site 104

.

Our crystallization trials included similar matrixes as the ones used for the double

A-site model, and resulted in the emergence of large (150-350 μm) crystals from rRNA

complexes with Apramycin (control), NB54, G418, NB84, NB74 and NB124 (Figure

4.13). All complexes were crystallized with a 1:1 A-site:AG ratio. The control

experimentations with Apramycin resulted in the emergence of well-ordered cubic-

shaped crystals that diffracted up to 2.6 Å resolutions in a home-source X-ray machine.

The unit cell parameters as well as crystal symmetry and structure solution statistics are

summarized in Table 4.6. The resulting structure indicated the binding of a single

Apramycin molecule to the A-site, which is in the ‘OFF’ state conformation (similarly to

the conformation observed upon Apramycin to the double A-site model) – Figure 4.14.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 75: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

63

Figure 4.13. Crystals emerged from single eukaryotic A-site rRNA duplex in complex with Apramycin

)A(, NB54 (B), G418 (C), NB84 (D), NB74 (E) and NB124 (F).

Table 3.8. Crystal data, data

collection and data refinement

statistics

Figure 4.14. Apramycin bound to

the extended single A-site model.

A. A-site 2D representation (‘OFF’

state). B. The three dimensional

structure determined with

Apramycin bound to the ‘OFF’

state with the conserved adenine

residues highlighted in green

(A1491), blue (A1492) and red

(A1493) and Apramycin in yellow.

C.2Fo-Fc electron density maps

contoured at 1.0σ around the

ligand and the rRNA residues.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 76: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

64

Table 4.6. Crystal data, data collection and data refinement statistics

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the observed and calculated structure factor

amplitudes, respectively. d Calculated using a random set containing 5% of observations that were not included in refinement.

Apra-single-cyt

Single A-site model -

Apramycin Crystallization Conditions

- Cryo-Protectant

Crystal Data P212121 Space group

a=28.62

b=36.94

c=87.64

α=90°

β=90°

γ=90°

Unit cell parameters (Å/°)

1 Za

Data Collection

Rigaku Raxis IV++

Tel Aviv university

Beamline

1.541 Wavelength (Å)

43.8-2.6 Resolution (Å)

14,526 Observed reflections

3,143 Unique reflections

100 Completeness (%)

100 in outer shell (%)

6 Rmergeb (%)

10 in outer shell (%)

14 Mean ((I)/sd(I))

2.5 in outer shell (%)

Structure Refinement

43.8-2.6 Resolution range (Å)

1,545 Reflections

19.15/23.25 R-factorc / Rfree

d (%)

698 Number of RNA atoms

1 Number of ligand molecules

- Number of water molecules

R.M.S.D

0.005 Bond length (Å)

0.879 Bond angles (°)

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 77: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

65

Nevertheless, the crystals emerged from complexation of the rRNA model with

the 5 read-through inducers poorly diffracted to 3.5 Å at best. Crystal analysis revealed

that all crystals had a P65 symmetry with unit cell parameters of a=49.19, b=49.19,

c=127.33, α=β=90°, γ=120°. Structure phases were determined using molecular

replacement with the rRNA residues of the Apramycin structure. Two molecules were

observed in the asymmetric unit. The molecules in an asymmetric unit were rotated in

60° facing one another (Figure 4.15 A), with the “sticky” ends resulting in the generation

of long rRNA fibrils that fabricated an ‘X’ shaped crystal packing (Figure 4.15 B).

Unfortunately due to the low resolution and the high flexibility of the conserved adenine

residues (A1491-3) it was impossible to model these residues and the absolute A-site

conformation could not be determined. In addition no clear indication was observed in

the electron density for the ligand’s presence. The R/Rfree values for the semi refined

structure were 30/35%, respectively.

Figure 4.15. Asymmetric unit )A( and crystal packing (B) of crystals emerged of complexes of the read-

through inducers NB54, NB74, NB84, NB124 and G418 with the single A-site model. The two molecules

present in the asymmetric unit were colored in blue and red for molecule 1 and 2, respectively.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 78: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

66

4.1.3 Crystal Structure Determination of 80S Yeast Ribosomes in Complex with AG

In addition to the single human A-site model used in our experimentations,

crystallization trials were also performed using complete 80S yeast ribosomes. Yeast 80S

ribosomes share great similarity with human ribosomes 99, 100

, with 100% sequence

similarity of the ribosomal decoding site. Ribosome purification and crystallization

techniques are fully described in the materials and methods section. In general, two

methods were used for complex generation: co-crystallization with up to 20 fold AG

excess and soaking of vacant ribosomes with up to 100 fold AG excess. Two synthetic

derivatives were used for complex generation: NB33, a strong inhibitor of human

ribosome translation that lacks any read-through activity 92

and NB127, a lead compound

developed in our lab that exhibited highest read-through activity thus far and has a great

selectivity to eukaryotic ribosomes 91

(Figure 1.7).

Our crystallization experiments resulted in the emergence of hundreds of

medium-large sized crystals (~100 μm). The initial crystals diffracted to ~12 Å (Figure

4.16 A, B). Crystals' quality was improved by systematic dehydration process that

resulted in resolution enhancement of up to 3 Å. Out of dozens of crystals scanned, we

collected 4 full data sets at 3.5-6 Å resolutions, two data sets for each compound, one

soaked and one co-crystallized (Table 4.7). Due to high radiation damage observed upon

crystals X-ray exposure, each data set was composed of a few merged data sets collected

from several crystals or several spots on the same crystal (Table 4.7). The asymmetric

unit contained two ribosomes with unit cell parameters that are identical to the ones

obtained in previously reported vacant yeast 80S ribosome structures 99, 100

.

Unfortunately, no density indicating the presence of bound AG could be identified.

A thorough investigation of the resulting crystal structures indicated two major

issues that might give an explanation to the lack of AG presence within their putative

binding site. One such topic was that in order to achieve structural homogeneity among

empty ribosomes purified for crystallization, yeasts were grown under glucose starvation.

Such conditions were previously reported to result in highly homogenous 80S ribosomes,

all exhibiting the same translational stage 134

. Apparently, such stress conditions resulted

in the expression of STM1, a protein that causes ribosome arrest upon stress conditions

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 79: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

67

135. Our X-ray results indeed indicated the presence of a large extra electron density in the

crystal structure. Mass Spectra experiments performed by Ben-Shem at al. 100

indeed

indicate the present of a non-ribosomal protein within the crystals. The protein was later

identified as STM1 and localization of its amino-acids within the extra electron density

revealed a rather tight interactions of STM1 with both ribosomal subunits (Figure 4.16

C); blocking the m-RNA entrance tunnel and forcing the ribosome into a ratcheted form,

preventing it from binding its substrates. This inhibition of dynamics appears to be the

source of translational arrest. STM1 presence also seemed to affect the A-site

conformation, resulting in a distorted conformation that changes the A-site’s geometry,

therefore preventing the AG molecules from binding to their putative binding site

(Figure 4.16 D, E).

Figure 4.16. Crystallization of yeast 80S ribosomes. A. Apo crystals emerged from crystallization trials. B.

Frozen crystal on a cryo-loop during X-ray analysis (SLS). C. 80S ribosome structure with STM1 highlighted in

red and the A-site in yellow. D.A closer glance at the ribosomal A-site, with conserved residues highlighted in

green (A1491), blue (A1492), red (A1493) and orange (G1408). Non canonical interactions between A1493 and

G1408 are highlighted in dashed blue lines. E. Superposition of eukaryotic A-sites from 80S yeast ribosome

structure (yellow) and 40S tetrahymena structure (cyan). F. Binding of Os-Hex to the ribosomal A-site. The

conserved Adenine residues are highlighted as in D.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 80: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

68

Table 4.7. Crystal data, data collection and data refinement statistics

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the observed and calculated structure factor amplitudes, respectively.

d Calculated using a random set containing 2% of observations that were not included in refinement.

NB127- Ribo-cox NB127- Ribo-soak NB33-Ribo-cox NB33-Ribo-soak

Ribosome:NB127 (1:20) Vacant ribosomes Ribosome:NB33 (1:20) Vacant ribosomes Crystallization Conditions

- NB127 100 fold (24h) - NB33 20 fold (24 h) Soak

Crystal Data P21 P21 P21 P21 Space group a=285.45

b=302.47

c=433.32

α=90°

β=98.85°

γ=90°

a=287.82

b=306.99

c=437.54

α=90°

β=99.07°

γ=90°

a=285.78

b=303.84

c=434.74

α=90°

β=99.07°

γ=90°

a=297.68

b=283.06

c=432.44

α=90°

β=97.87°

γ=90°

Unit cell parameters (Å/°)

2 2 2 2 Za

Data Collection PXI - X06SA, SLS

Villigen, Switzerland

PXI - X06SA, SLS

Villigen, Switzerland

PXI - X06SA, SLS

Villigen, Switzerland

PXI - X06SA, SLS

Villigen, Switzerland Beamline

1.00 1.00 1.00 1.00 Wavelength (Å) 7 3 3 5 No. of datasets merged 200-3.5 300-4.2 300-4.0 400-6.0 Resolution (Å) 4,647,856 1,510,820 792,785 888,919 Observed reflections 939,466 464,259 191,313 175,94 Unique reflections 99.8 99.9 99.1 99.9 Completeness (%) 100 100 98.7 99.9 in outer shell (%) 18.9 18.3 20.5 21.6 Rmerge

b (%)

51.8 42.8 57.6 46.4 in outer shell (%) 4.8 6.1 5.93 5.5 Mean ((I)/sd(I)) 2.97 2.85 2.67 2.6 in outer shell (%)

Structure Refinement 56.7-2.8 300-4.2 300-4.0 293-6.0 Resolution range (Å) 2,719 4,206 3,214 173,340 Reflections 18.1/22.8 19.1/22.9 18.2/22.3 20.5/23.7 R-factor

c / Rfree

d (%)

R.M.S.D 0.013 0.015 0.015 0.016 Bond length (Å) 1.512 1.672 1.651 1.713 Bond angles (°)

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 81: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

69

In addition, as indicated, the initial crystals obtained were soft and fragile, and

therefore poorly diffracted to 12 Å and less. These parameters indicated that the water

content within the crystals was too high; therefore several dehydration steps were used to

lower water content by replacing the water molecules with organic solvent (PEG) and the

addition of poly-amines (Os-hex/Co-hex) to decrease RNA repulsion and tighten crystal

packing; thus improving crystals diffraction properties. However, despite the large excess

of AG in the crystallization and dehydration steps, 2 Os-Hex molecules were shown to

occupy the ribosomal A-sites in all tested crystals; therefore preventing the AG from

binding their putative binding site (Figure 4.16 F).

4.1.4 Leishmania A-site Crystal Structure Determination

In the absence of a solid evidence for AG binding to models mimicking their

binding sites in human and in continuation of our experimentations with the double A-

site model, we generated a double A-site construct containing a single mutation in

comparison to the human A-site. Such mutation was meant to limit the flexibility of

A1491 by generating a Watson-Crick pair with position 1409 (C1409U). Such

interactions were meant to stabilize the geometrical conformation promoting ligand

binding; thus increasing our chances to get strong indications for ligand binding under

physiological conditions. Interestingly, introduction of this single mutation resulted in an

A-site that is identical to the A-site of the eukaryotic parasite, Leishmania, (Figure 4.17)

known to cause mild to severe symptoms in humans, as well as dogs; affecting as many

as 12 million people worldwide, with 1.5–2 million new cases each year.

Figure 4.17. Secondary structures of the

bacterial (A), leishmanial (B) and human

(C) cytoplasmic A-sites. A/G1408 -

orange, A1491 - green, A1492 - blue and

A1493 - red. The rRNA residues are

numbered according to the numbering

used in E.coli 16S rRNA.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 82: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

70

Our structural investigations resulted in the crystal emergence and determination

of the three dimensional structures of two AG representatives, G418 and Apramycin

(Figure 1.1), bound to leishmanian rRNA double A-site duplexes model at 2.65 Å and

1.4 Å resolutions, respectively (G418-Leish, Apra-Leish; Figures 4.18, 4.19, 4.20 Table

4.8). The resulting structures indeed demonstrated a high geometrical similarity between

the leishmanial A-site and the bacterial A-site, despite the 33% dissimilarity in their

nucleotides sequence (5 out of 15 nucleotides located in positions 1408-1410 and 1490-

1491 are different; Figure 4.17). These geometric similarities do not come as a surprise,

since both sites are believed to be an important molecular switch dictating the tRNA

recognition at the ribosomal A-site. However, since AG mostly interact with the

nucleotide bases, and there are significant differences in sequence, we hypothesized that

the obtained interaction patterns and conformational changes induced by different AG

might differ in Leishmania from the ones present in bacteria.

Overall, in both the G418-Leish and Apra-Leish structures a single AG molecule

was found to specifically interact with the deep/major groove of each A-site (Figure

4.19). In the Apra-Leish structure 2 additional Apramycin molecules were found to

interact with the G-C pairs region connecting the two putative binding sites present in the

rRNA model (Figure 4.19 B, Figure 4.21). Similar non-specific interactions with the

non-natural constructed G-C pair region were previously reported in our experiments.

However, despite the fact that both ligands (G418 and Apramycin) were shown to bind

the same region of their primary binding site, the two ligands demonstrated a

significantly different interaction pattern with the rRNA construct, and thus each AG

induced a different conformational change to the A-site. The conformational change

induced by G418 is similar to that already observed for the bacterial A-site, where two

conserved adenine residues, A1492 and A1493, are fully bulged out from the helical core

(Figure 4.22). In contrast, Apramycin induces a rather different conformation, in which

the A1492 base moiety is sequestered within the helical core, forming a pair with G1408,

while A1493 is bulged out (Figure 4.19, Figure 4.22).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 83: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

71

Figure 4.19. 2D (right panels) and 3D (left panels) representations of the double A-site complexes as

obtained in the X-ray crystal structures. A. The double A-site construct in complex with G418 (PDB ID:

4K32): each A-site binds one molecule of G418 (yellow). Both A-sites are in the 'ON' state conformation

where A1492 (blue) and A1493 (red) are bulged out from the helical core. B. The double A-site construct

in complex with apramycin (PDB ID: 4K31): each A-site binds one molecule of apramycin (yellow). Two

additional apramycin molecules can be observed in the G-C pairs region separating between the two A-sites

(marked in black arrows). Both A-sites are in the 'OFF' state conformation where A1493 (red) is bulged out

from the helical core and A1492 (blue) interacts with G1408 (orange) in a non-canonical manner. A1491

(green) forms a Watson-Crick pair with U1409 in both complexes.

Figure 4.18. Crystals emerged from

crystallization experiments of complexes of

the leishmanial A-site model with G418 (A)

and Apramycin (B).

A. B.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 84: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

72

Figure 4.20. Electron density maps of the Apra-Leish (A) and G418-Leish (B) structures. 2Fo-Fc maps are

contoured at 1.0σ around rRNA molecules (cyan) and 1.5σ around ligand molecules (dark blue).

Figure 4.21. Surface representation of apramycin bound to the double A-site rRNA construct corresponding to its

putative binding site in leishmanial ribosomes. Apramycin molecules are represented as yellow sticks. The surface

colors indicate residue conservation among prokaryotic and eukaryotic systems, where residues marked in cyan

are highly conserved among all kingdoms, residues marked in light pink are rather diverse and residues marked in

light yellow are non-A-site nucleotides that were used to stabilize the model. Double helices (left to right) are

rotated by 90° around the helical core. Apramycin molecules are numbered I-IV (top to bottom).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 85: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

73

Table 4.8. Crystal data, data collection and data refinement statistics

a Number of RNA molecules in the asymmetric unit.

b Rmerge = 100 x Σhklj|Ihklj-<Ihklj>|/ Σhklj<Ihklj>.

c R-factor = 100 x Σ||Fo| - |Fc|| / Σ|Fo|, where |Fo| and |Fc| are the observed and calculated structure factor

amplitudes, respectively. d Calculated using a random set containing 5% of observations that were not included in refinement.

Apra-Leish G418-Leish

4K31 4K32 PDB code

Crystal Data P31 P21212 Space group

a=b=32.79

c=107.40

a=33.08

b=90.76

c=47.01

Unit cell parameters (Å)

1 1 Za

Data Collection

ID14-4 ESRF ID14-4 ESRF Beamline

0.9394 0.9394 Wavelength (Å)

35.8-1.4 23.5-2.65 Resolution (Å)

67,545 18,136 Observed reflections

23,324 3,615 Unique reflections

94.5 80.9 Completeness (%)

98.0 84.5 in outer shell (%)

4.8 4.8 Rmergeb (%)

13.7 30.8 in outer shell (%)

14.1 25.11 Mean ((I)/sd(I))

5.7 5.15 in outer shell (%)

Structure Refinement

35.8-1.4 23.5-2.65 Resolution range (Å)

22,089 3,612 Unique reflections

22.6/26.4 19.5/23.9 R-factorc / Rfree

d (%)

940 955 Number of RNA atoms

4 2 Number of ligand molecules

209 0 Number of water molecules

R.M.S.D

0.025 0.013 Bond length (Å)

2.47 2.162 Bond angles (°)

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 86: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

74

Figure 4.22. Crystal structure visualization of (A) G418 or (B) Apramycin (yellow) - leishmanial A-site

rRNA complexes. Electron density 2Fo-Fc maps are contoured at 1.0σ around the AG. (C) A representation

of Apramycin bound to the human A-site construct (PDB code 2G5K). G1491 is highlighted in green,

A1492 in blue, A1493 in red and U1409 in purple. (D) Superimposition of G418 bound to the bacterial A-

site (orange) and the leishmanial A-site (blue). G418 is highlighted in ball-and stick representation.

Superimposition was performed using the PyMol software align algorithm using all identical atoms

(r.m.s.d. 0.9 Å). PDB codes are 1MWL and 4K32 for the bacterial and Leishmania structures, respectively.

(E) Superimposition of Apramycin (ball and stick) bound to the bacterial (orange) and leishmanial (blue)

A-sites (r.m.s.d. 2.2 Å). PDB codes are 1YRJ and 4K31 for the bacterial and Leishmania structures,

respectively. (F) Superimposition of Apramycin (ball and stick) bound to the human cytoplasmic (green)

and leishmanial (blue) ribosomal A-sites (r.m.s.d. 1.4 Å). PDB codes are 2G5K and 4K31 for the human

and Leishmania structures, respectively. In all figures A/G1491, A1492, A1493 and A/G1408 are

highlighted in the relevant colors.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 87: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

75

4.1.4.1 The Binding Mode of G418 to the Leishmanial A-site - ‘ON’ State.

Upon binding to the deep major groove of the leishmanial A-site, G418 forms 14

hydrogen bonds with the rRNA bases and phosphate oxygen atoms (Figure 4.23). Ring I

forms a pseudo base pair with G1408, by forming 2 hydrogen bonds involving the ring

oxygen and the 6’-OH group (Figure 4.23 B). The planar nature of these interactions is

dominated by stacking interactions of ring I and A1491 (Figure 4.23 C). On the opposite

side of ring I, two OH groups located at the 3’ and 4’ positions make additional contacts

with the oxygen atoms belonging to the phosphate backbone of the two conserved

adenine residues, A1492 and A1493 (Figure 4.23 D). These interactions prevent the two

adenine residues from flipping back inside the helical core. This interaction is therefore

deemed to be highly important for the induction of the flipped out conformation of

A1492 and A1493. The chiral exocyclic methyl group at 6' position, (R)-6'-Me, points in

the middle of the A1491-U1409 pair (Figure 4.23 D); thus adding some hydrophobic

stabilization as the one observed upon G418 ring I interaction with the G1491-C1409 pair

in bacteria 114

. Ring II interacts with three sequential residues A1493, G1494 and U1495

(Figure 4.23 E). Due to the high conservation of these residues, similar interactions were

previously highlighted in the exploration of w.t. and mutant bacterial A-sites in complex

with AG containing a 2-DOS ring 114, 137

. Ring III, forms 6 hydrogen bonds with G1405,

U1406 and C1407, these interaction are typical to 4,6-disubstituted AG containing a

garoseamine ring (ring III) at position 6 (Figure 4.23 F). Similar interactions were

previously reported for the interaction of 4,6- disubstituted garoseamine derivatives such

as Gentamicin C1A and G418 with bacterial A-sites 110, 114

.

Superposition of the G418-Leish structure with the crystal structure of G418 bound to the

bacterial A-site (G418-Bact) indicated a rather similar binding mode with r.m.s.d values

of 0.9 Å (Figure 4.22 D). The overall conformation of the binding site upon G418

binding was nearly identical to the one present in the bacterial A-site, where the two

conserved adenines 1492 and 1493 are flipped out from the helical core prone to interact

with both the mRNA and the tRNA codon:anti-codon complex. This conformational state

was earlier referred to as an ‘ON’ state 19, 121

and is known to mimic the conformational

change resulting from the complexation of a cognate aminoacylated-tRNA molecule and

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 88: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

76

the mRNA at the bacterial ribosomal A-site; thus signaling for the continuation of

translation process. The induced conformational change upon AG binding to bacterial

ribosomes is believed to play a major role in their ability to induce translational

miscoding 110

. Although no direct evidence is currently available showing that G418

induces miscoding by leishmanial ribosomes, G418 has long been marked as a potent

miscoding agent in eukaryotes 24

. In addition, recent work has demonstrated the ability of

a structurally related derivative, Paromomycin, to induce miscoding in Leishmania 138

.

The structural similarity between the two structures (G418-Leish and G418-bact) together

with documented evidence for AG such as Paromomycin enhancing miscoding events in

leishmanial ribosomes might suggest a similar mechanism of action in Leishmania.

Figure 4.23. Description of the contacts between G418 and the leishmanial A-site. (A) The 3D structure of

bound G418. Ring numbers (I-III) and atom names are specified. rRNA atoms are numbered according to

the E. coli numbering. Hydrogen bonds and salt bridges are presented as black dashed lines. Bond lengths

are presented in dark green in ångström - Å. (B-F) The atomic details of the contacts involving the rings

with conserved and non-conserved rRNA residues.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 89: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

77

4.1.4.2. The Binding Mode of Apramycin to the Leishmanial A-site - ‘OFF’ State.

In contrast to G418, Apramycin demonstrated a looser interaction network,

maintaining only 6 direct contacts with the leishmanial A-site (Figure 4.24). Four of

these contacts are mediated by ring II (2-DOS) that interacts with the RNA bases and

phosphate backbones of the 3 conserved residues G1405, U1406 and G1494 (Figure 4.24

B). The 2 additional remaining bonds are between the 2’-NH2 group located at the

bicyclic ring I to the O6 atoms of G1494 and G1408 (Figure 4.24 A). As can be seen

from the superposition with the bacterial-A-site-Apramycin complex (Apra-Bact)

(Figure 4.22 E) the binding pattern of Apramycin to the leishmanial A-site is rather

distinct from the one observed in bacteria (r.m.s.d 2.2 Å). As a matter of fact, when only

the two ligand molecules are compared their atoms barely overlap; with the molecules

oriented in a ~180° horizontal flip one to another. As a result Apramycin induces a

distinctive conformational change upon binding to the leishmanial A-site; where only

A1493 is flipped out from the helical core. A1492, which is directed towards the helical

core forms pseudo canonical interactions with G1408 (Figure 4.24 C). A conformation

where one of the two conserved adenines (A1492 and A1493) or both are directed

towards the helical core was earlier referred as an ‘OFF’ state conformation 121

. This

conformation usually occurs when an inappropriate or no tRNA molecule is present at the

ribosomal A-site; therefore it might signal for translational arrest. Despite the great

dissimilarity of the Apramycin structure to the previously reported X-ray structures, our

results are in good agreement with a recent solution NMR structure indicating the lack of

A1492 and A1493 destacking upon Apramycin’s binding to an rRNA bacterial A-site

model 106

. The obtained results also correlate with some recent evidence for Apramycin’s

ability to inhibit translocation in bacterial ribosomes, but not to induce bacterial

miscoding 106, 139

or PTC suppression in eukaryotes 92

.

Interestingly, the binding pattern observed upon Apramycin binding to the

leishmanial A-site was found to be rather similar to the one observed in our previous

complexes of Apramycin with the human cytoplasmic A-site constructs (superposition

r.m.s.d 1.4 Å; Figure 4.22 C, F). The human A-site departs from the leishmanial one by

a single nucleotide located in position 1409 (C1409 human vs. U1409 Leishmania -

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 90: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

78

Figure 4.17). However, although these two A-sites share greater sequential similarity

when compared to the bacterial one, the geometry of the two A-sites is more distinct due

to the abrogation of the 1409:1491 Watson-Crick pair in the human A-site; giving its

helical core a more complex conformational arsenal. Nevertheless, despite the noticeable

differences in the orientations of A1491-A1493, Apramycin binding and interaction with

the leishmanial A-site is nearly identical to the one observed in the human A-site (Figure

4.22 F). The obtained results indicate a rather conserved interaction pattern in eukaryotes.

Such similarity could be attributed to the presence of the highly conserved guanine

residue in position 1408, maintaining a preserved interaction with the OH moiety located

in the bicyclic ring I (Figure 4.24 A).

Figure 4.24. Description of the contacts between Apramycin and the leishmanial A-site. Ring numbers (I-

III) and atom names are specified. rRNA atoms are numbered according to the E.coli numbering. Hydrogen

bonds and salt bridges are presented as black dashed lines. Bond lengths are presented in dark green in

angström - Å. The atomic details of the contacts involving ring I (A) and the bicyclic ring II (B) with

conserved and non-conserved rRNA residues. (C) Pseudo Watson-Crick interactions of A1492 and G1408.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 91: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

79

4.1.5. In-vitro Inhibition of L. donovani and L. major Promastigote Growth by AG.

The structural evidence obtained in this study increases our understanding of the

molecular mechanisms of AG interaction with leishmanial ribosomes. However, to date a

comprehensive study comparing susceptibility to different AG derivatives was not carried

out. In order to evaluate the effect of AG on Leishmania growth 5 representative

derivatives were chosen. Paromomycin and Neomycin B were chosen as representatives

of the 4,5-disubstituted 2-DOS group; G418 and Gentamicin as 4,6-disubstituted and

Apramycin as mono-substituted. The representatives of each group differ by the nature of

the substituent group at the 6’-position of ring I (Figure 1.1), which is known to be an

important component of selectivity towards eukaryotic species 140

. Drug susceptibility

was tested using two species, L. major and L. donovani, which induce cutaneous and

visceral leishmaniasis in humans, respectively. The concentrations giving 50% of growth

inhibition (IC50) are listed in Table 4.9. All AG tested inhibited Leishmania growth in a

dose dependant manner.

The IC50’s obtained for the 6’-OH AG (Paromomycin and G418) were in the μM

range, and showed good agreement with previous published values for these compounds

138, 141, 142. The results were also similar to the MIC values reported in bacterial systems

for the two compounds 85, 88, 143

; thus adding additional support to the suggested

similarity in their mechanism of action in bacteria and Leishmania. However, the

obtained results indicate that G418 is ~10 times more potent than Paromomycin against

the two Leishmania strains tested. Although no such potency gap for the two derivatives

has been documented in w.t. bacterial strains, similar tendency has been reported in

A1408G mutant bacteria 114

, sharing greater binding site similarity with Leishmania.

These results also correlate well with the higher miscoding 24

and PTC suppressing

potency reported for G418 over Paromomycin in eukaryotes 91

. The obtained IC50 values

for the 6’-NH2 derivatives (Neomycin and Gentamicin) were higher than those obtained

for the 6’-OH derivatives, and are also in good agreement with previously reported work

demonstrating the lower potency of Neomycin B, when compared to Paromomycin, for

treatment of leishmaniasis 138

. These results are also correlated well with the facts that 6’-

NH2 derivatives are less effective against AG resistant bacterial strains containing an

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 92: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

80

A1408G substitution, and have a very limited read-through and misreading activity in

higher eukaryotes.

However, unlike in bacterial systems, where Apramycin is conserved as a potent

antibacterial agent, our IC50 results in Leishmania indicated a rather low activity of the

compound (IC50>2,000 μM; Table 4.9). These results could be explained either by the

distinct A-site conformation observed in X-ray experiments designed to explore

Apramycin’s binding pattern in bacteria 139, 144

(Figure 4.22 E), or by the possibility of

alternative binding sites at the bacterial ribosome. The conformational alternatives

suggested for Apramycin in bacterial systems cannot be obtained in the leishmanial A-

site due to alternations in the rRNA sequence preventing several H-bonds from forming;

therefore limiting the binding affinity of such conformations. The recent indications of

the rather low translation inhibition of human ribosomes by Apramycin 115

, altogether

with the structural similarity in Apramycin’s binding to eukaryotic A-sites further support

our bio-activity results indicating a low susceptibility of eukaryotic parasites to

Apramycin.

Table 4.9. In-vitro inhibition of Leishmania promastigotes by natural AG

IC50 (μM)

L.donovani L.major

48.1 ± 5.8, n=6a 31.4 ± 5.7, n=5

a Paromomycin

>2000 >1000 Neomycin

5.8 ± 0.9, n=4a 1.95 ± 0.05, n=2

a G418

>2000 273.1 Gentamicin

>2000 >1000 Apramycin a Each value represents the mean ± standard error of n repeats.

The obtained susceptibility results for the natural AG highlighted the superiority

of the 6'-OH di-substituted derivatives for the treatment of leishmaniasis. Adding these

results to the similar tendency of the tested AG to induce translational read-through

activities in human encouraged us to further test the leishmanial parasites susceptibility to

our lead NB compounds (NB74, NB 84 and NB 124, Figure 1.7). These compounds

were shown to induce the highest read-through activities in eukaryotes reported for

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 93: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

81

synthetic derivatives thus far 88, 91

; and indeed, our susceptibility tests indicate that the

NB compounds were able to inhibit leishmanial growth at the low micromolar range

(Table 4.10); with NB124 exhibiting the highest anti-leishmanial activity values. The

IC50 values were 3-6 fold lower than those obtained for paromomycin, which was used as

a basic scaffold for the design of our NB compounds, but 2.5-5 times higher than those

observed for G418, of which rings I and II are identical. The improved activity of our NB

compounds when compared to Paromomycin might be a direct effect of the 6'-(R)-

Methyl group presence in the NB compounds directly interacting with the π systems of

the A1491-U1409 pair as observed in the G418 structure (Figure 4.23 D). In addition,

the presence of the 5''-NH2 group when compared to the 5''-OH in paromomycin, might

add some additional stabilization with the G1408 residue (Figure 4.25).

Table 4.10. In-vitro inhibition of Leishmania promastigotes by synthetic AG and toxicity measurements in

HEK293 cells (LC50)

Cell Toxicity

LC50 HEK293 (mM)b

Anti-leishmanial activity

IC50 L.major (μM)a

1.31 ± 0.06 1.95 ± 0.05 G418

4.13 ± 0.51 31.4 ± 5.7 Paromomycin

22.7 ± 1.06 8.2 ± 2.1 NB74

5.77 ± 0.68 10.4 ± 1.8 NB84

5.40 ± 0.45 5.6 ± 0.9 NB124 a Each value represents the mean ± standard error of at least 2 repeats performed in triplicates.

b The LC50 values were previously reported [

88, 91].

In addition to exhibiting some excellent anti-leishmanial properties the developed

NB compounds were also demonstrated to be less toxic than both G418 and

Paromomycin, as indicated previously in our cell toxicity assays (Table 4.10) 88, 91

. These

preliminary results are highly encouraging and gave rise to some additional trials

currently ongoing in our lab.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 94: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

82

Figure 4.25. Surface representations of AG binding sites in leishmanial ribosomes. (A) The G418 position

as crystallographically determined and putative binding modes of Paromomycin (B) and NB124 (C) as

obtained by manual docking based on this ligands binding to the bacterial ribosome. AG are represented in

stick representation and highlighted in yellow. The coloring of the surface represent residue conservation

among prokaryotic and eukaryotic systems, where residues marked in cyan are highly conserved among all

kingdoms and residues marked in light pink are rather diverse. The Paromomycin and NB124 positions

were obtained by superimposition of the A-site coordinates from the G418/leishmanial A-site structure

(PDB ID: 4K32) and the paromomycin coordinates from the crystal structure of paromomycin bound to the

bacterial A-site (PDB ID: 1J7T ). Possible H-bonds are represented in dashed yellow lines. H-bonds

alternatives for the 5''-NH2 group of NB124 are highlighted in a red circle. Figure was made using Pymol.

(D) A 2-D representation of the additional contacts of 5''-NH2 group with G1408.

4.1.6. Aminoglycosides Mechanisms of Action in Leishmania

The combination of our structural and biochemical data shows for the first time the

mechanistic parameters governing the interactions of AG in leishmanial ribosomes, and

supplies the molecular explanation for their potent activity against Leishmania. The data

also reveals that in contrast to bacterial systems, the substitution pattern of AG around the

2-DOS ring affects their binding mode and might induce an entirely different

conformational change upon binding. Our data also indicate that the induction of an ‘ON’

A. B.

C.

A1491

A1491

A1491

G1408

G1408

G1408

N

NNH

N

O

N

OH

H

H

H

+H2N

O

H

5''-Ring III

Ring I

6'

ring oxigen

NB compound

G1408

D.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 95: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

83

state conformation is highly important for AG potency as anti-Leishmania agents; thus

implying the significance of miscoding events in the killing mechanism. The data

highlights the importance of 6'-OH ring I for the induction of an 'ON' state conformation.

Based on the obtained data we could suggest a simple diagram for AG mechanism of

action against leishmanial cells (Figure 4.26).

Figure 4.26. General scheme of AG activities in Leishmania. Normal translation is presented in the middle

where cognate tRNA is highlighted in green. The lower part describes mistranslational events in the present

of 6'-OH AG inducing and 'ON' state conformation upon binding (6'-OH AG are highlighted in red). The

upper part describes a simple inhibition of translation upon Apramycin's binding (Apramycin is highlighted

in yellow).

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 96: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

84

4.2. The Development and Employment of a Highly Sensitive Immunoassay for the

Detection of AG in Biological Samples

Our crystallization trials increased our understanding of how AG act in

eukaryotes and gave rise to the further development of new and improved derivatives.

The promising results of our pre-developed NB compounds in the treatment of PTC

induced genetic disorder altogether with our recent results in Leishmania necessitated a

fast and effective methodology for the detection and quantification of the developed lead

NB-compounds in various biological derived matrices; such methodology will be of high

importance in further progress in drug development.

Over the last few decades many analytical and bioanalytical assays were

suggested for the qualitative and quantitative analysis of AG that are in clinical,

veterinary and agricultural use to treat bacterial infections. These methods were applied

for the detection of drug residues in a wide range of biological derived matrices such as

cell tissues 132, 145-147

, serum 148-154

, milk 145, 148-152, 154-160

, eggs 147, 157

, and honey 157

.

Chemical methodologies included the application of gas chromatography (GC) 161

, TLC

162, high-performance liquid chromatography (HPLC)

163 and capillary electrophoresis

(CE) 164

. Biological methodologies included the development of microbiological assays

165, radiochemical and radioimmunochemical assays (RIA)

166, enzyme linked- and

flouro- immunoassays (ELISA, FIA) 149, 167, 168

, nano-sensors and nano-particle based

immunoassays 169, 170

.

The main advantages of using chemical methods for the detection of AG in

biological samples is the possibility of detecting a wide variety of therapeutic agents

simultaneously altogether with monitoring their corresponding metabolitic products.

Nevertheless, due to their high polarity and the lack of chromophoric or florophoric

properties AG chemical detection is quite a challenging task. Recent work have

demonstrated the use of various analytical methodologies conjugated to MS-MS detectors

171, 172, whereas other works used various UV/floroscentic derivatization methods that

were further exploited for the detection of the derivatizied products 173

. These methods

were found to be highly sensitive and demonstrated an efficient procedure for the

detection of AG. However, applying these methods on biological derived matrices

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 97: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

85

required several pre-purification and sample concentration steps prior to detection

process. Moreover, these methods cannot be readily utilized in all laboratories and

engage the use of specific instrumentation which is highly costly and requires well

trained human resources to operate it and analyze the resulting data.

Immuno-based methodologies such as ELISA, FIA and RIA have shown to be as

sensitive and accurate as the chemical methods 174

. These methods could be easily

applied on a wide variety of biologically derived matrices, are simple to perform and

analyze, and do not require the use of high cost instrumentations. Immuno-based

methodologies are commercially available for the detection of some widely used natural

AG such as gentamicin, streptomycin, dihydrostreptomycin, neomycin and kanamycins

(eg. MaxSignal® gentamicin ELISA Test Kit, AG enzyme immune-assay-EIA kit for the

detection of gentamicin, neomycin, streptomycin and dihydrostreptomycin -

Europroxima, and kanamycins ELISA kit - Wanger). These commercially available kits

can be exploited for highly sensitive detection of AG in various matrices, including body

fluids, cell tissues and food products. Similar immuno-based methodologies are used in

hospitals to monitor the serum levels of clinically used AG such as gentamicin and

amikacin. However, these assays are mostly used for the detection of AG antibiotics that

are based on a neamine core, containing an amino group at their 6' position (ring I,

Figure 4.27). The use of the derived antibodies for the detection of therapeutic

derivatives containing other substituents on the neamine core is quite limited due to low

cross reactivity values. Recent documentation demonstrated the superiority of 6'-

hydroxyl containing AG such as paromomycin and G418 over the 6'-amino derivatives in

the treatment of various genetic disorders 26

. These derivatives are mostly based on either

a paromamine or an NB82 2-ring core (Figure 4.27) and are extensively explored for the

treatment of various genetic diseases 82, 85, 88, 90

. To our knowledge, no immunoassays

exist for the detection of AG containing a 6'-hydroxyl group.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 98: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

86

Figure 4.27: Chemical structures of standard and semi-synthetic AG derivatives used in the study.

4.2.1 Preparation of Immunization Conjugate

The main goal of this part of the present study was to develop an

immunochemical assay for monitoring lead compounds of NB-series in biologically

derived samples. The specific aims included the production of a generic polyclonal

antiserum that will be able to recognize a wide variety of NB compounds (Figure 4.27)

that have previously been shown to exhibit great potential for the treatment of various

nonsense mutations related genetic diseases 34, 82, 83, 85, 87-91

.

6'-(R)-methyl-paromamine (NB82) (Figure 4.27), a semi-synthetic 2-ring

fragment derived from G418, is a common scaffold, appearing in nearly all compounds

of NB-series (Figure 4.27). We assumed that by choosing NB82 as hapten for

immunogen preparation we could obtain polyclonal antibodies that will be able to

recognize a diverse selection of these compounds. Similar methodology was previously

described by Van Amerongen and coworkers 158

by using neamine as an immunogenic

hapten for the generation of polyclonal antiserum capable of recognizing some natural

AG such as neomycin, kanamycin A and gentamicin.

Our next challenge was to choose a proper conjugation strategy for the

preparation of immunization conjugate. Since NB82 contains three primary amino

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 99: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

87

groups, it could be easily subjected to either direct conjugation with carrier proteins via

the formation of amide bonds using the EDC method 175

, or to indirect conjugation

methods using linking agents such as glutaraldehyde 132, 175

. These methods were

previously reported as highly efficient for the generation of anti-AG antibodies.

However, due to amino group multiplicity the coupling process usually results in a

spontaneous non-specific linkage to any amino group of the subjected hapten. Some of

our lead compounds, like NB84, contain an (S)-4-amino-2-hydroxybutanoyl (AHB)

moiety at their N-1 position (Figure 4.27). Our earlier studies have demonstrated that the

addition of AHB group selectively at N-1 position significantly enhances the read-

through activity of the resulted derivatives in comparison to that of the parent structure

without the AHB, while its toxicity profile was not significantly affected 85, 88, 90

. These

derivatives are rather bulky, and could interfere with the antibodies binding affinity

towards some of the ligands arsenal; therefore we assumed that the best linking option

would be through the N-1 position. Taking into account that the antibody raised against a

hapten linked to the carrier protein through N-1 position would have similar affinity to

both compounds with and without AHB, further supported our strategy for linking NB82

to a carrier protein selectively at N-1 position.

In order to selectively conjugate the NB82 scaffold to a carrier protein via the N-1

position we first had to selectively protect the additional two amino groups at positions 2’

and 3 to prevent them from reacting with the linking agent. This was done by selective

azidation of NB82 at 2’ and 3 positions to yield 2’,3-diazido-NB82 (compound A,

Figure 3.1). Treatment of compound A with glutaric anhydride afforded compound B in

62% isolated yield. The carboxylic acid function of compound B at its N-1 side-chain

was activated by reaction with N-hydroxysuccinimide in the presence of DCC to yield

compound C. Conjugation of the NHS-activated compound C with the carrier protein

KLH was followed by the conversion of two azido groups to the corresponding amines

using Staudinger reaction (Me3P, pH 9.2) to yield the immunization conjugate NB82-

KLH (compound E, Figure 3.1). The resulting conjugate was used as an immunization

agent and was injected to two rabbits.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 100: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

88

4.2.2 Development of an Indirect Competitive ELISA

The two rabbits immunized with the NB82-KLH conjugate showed high antibody

serum titers, as indicated in checkerboard homologous-assays performed using the NB82-

OVA conjugate as a coating antigen (Figures 4.28, 4.29). These experiments were

further used to determine the optimal concentrations of coating antigen (NB82-OVA,

1:1), rabbit antiserum (8245) and secondary antibody to be 1:8,000, 1:80,000 and

1:20,000, respectively. These conditions were used to determine the I50 value and assay’s

detection limit (I20) of NB82 to be 50±6 ng mL-1

and 8±1 ng mL-1

(n=10), respectively

(Figure 4.30, Table 4.11).

Figure 4.28: Antiserum activity of pre-immune (PI) rabbits, two weeks after 1st immunization with 1 mg

NB82-KLH conjugate (1st bleed), 2 weeks after 1

st boost with additional 1 mg immunogen (2

nd bleed), and

after 2nd

boost (3rd

bleed). Microtitter plate assay performed by using NB82–OVA conjugate 1:1 in 1:8,000

dilution and antisera of rabbits 8244 (A) and 8245 (B) diluted in a range of 1:5,000-1:40,000. Each bar

represents the mean of 3 independent repeats performed in duplicates.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 101: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

89

Figure 4.29: Antigen recognition (Chessboard) assay. Coating antigen (NB82-OVA, prepared in 1:1 molar

ratio of NB82 and OVA) dilutions were 1:8,000-1:64,000 in carbonate buffer pH 9.6; 3rd

bleed antiserum

derived from RB8244 (A) and RB8245 (B) dilutions were 1:5,000-1:160,000. Each bar represents the mean

of at least 3 independent repeats performed in duplicates.

Figure 4.30: Standard curve of NB82 (Homologous assay). NB82-OVA conjugate dilution 1:8,000;

Antisera dilution (Rabbit 8245) 1:80,000. I50 50±6 ng mL-1

; Limits of detection: 8±1 ng mL-1

. Each value

represents the mean±S.E.M. (Standard error mean) of 10 independent repeats performed in duplicates.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 102: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

90

Table 4.11: Concentrations at 50% inhibition (I50) and limits of detection (I20) of various AG using the

homologous ELISA with NB82-OVA as a coating antigena.

Compound I50

(µg/mL) Detection limit (I20) (µg/mL)

NB82 0.05±0.006 0.008±0.001

G418 0.4±0.04 0.06±0.02

NB125 1.25±0.14 0.3±0.03

Paromamine 3.0±0.34 0.6±0.1

NB84 3.1±0.75 0.7±0.1

NB74 3.5±0.15 0.7±0.1

NB124 12±1.1 3±0.7

Gentamicin 130±8.99 20±3

NB123 250±22.6 40±1

Paromomycin 325±56.6 50±2

NB122 400±34.2 80±4

Neomycin 700±71.7 90±4

Kanamycin A 1000±144 120±11

Neamine 2000±251 150±14 a

NB82-OVA conjugate dilution 1:8,000 was used as coating antigen; Antisera dilution (Rabbit 8245) 1:80,000. Each value represents

the mean±S.E.M. of at least 3 experiments performed in duplicates.

In order to evaluate the efficiency of the assay for the detection of various AG, 13

representative members were chosen to be detected for antiserum cross-reactivity via

homologous-cross-reactivity assays, using the NB82-OVA conjugate as a coating

antigen. Five of them including G418, gentamicin, paromomycin, neomycin and

kanamycin A are natural AG that are mostly known for their antibacterial activities

(Figure 4.27). Six compounds including NB74, NB84, NB122, NB123, NB124 and

NB125, are semi-synthetic agents systematically developed in our laboratory as read-

through inducers and have previously shown to bear a great potential of treating various

genetic disorders caused by premature stop codon mutations 33, 34, 82, 83, 85, 87-90

. Three

additional compounds selected in this study included 2-ring structures as common

scaffolds for various natural and semi-synthetic AG: neamine, a common scaffold of

gentamicin, neomycin B and kanamycin A; paromamine, a common scaffold of

paromomycin, NB122 and NB123; NB82, a common scaffold of G418, NB74, NB84,

NB124 and NB125. As can be seen from the data in Figure 4.31, antiserum antibodies

were able to recognize all tested members at various sensitivity ranges, where NB82, as

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 103: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

91

expected, demonstrated the lowest I50 value, thus indicating highest antibody affinity

(Table 4.11).

Earlier studies have shown that AG with a 6'-OH group on ring I (such as G418

and paromomycin, Figure 4.27) are more effective read-through inducers than those with

an amine at the same position 26

. The affinities of serum antibodies towards the

structurally related AG containing a secondary (R)-6'-OH on ring I (NB82, G418, NB74,

NB84, NB124, NB125) were higher than those obtained for similar compounds

containing either a primary 6'-OH (NB122, NB123, paromomycin, paromamine) or a 6'-

NH2 (neomycin, neamine, gentamicin, kanamycin A). These results correlate well with

our expectations that the resulting antibodies will show higher specificity towards

compounds that share greater similarity with the parent compound used for

immunization. Using NB82-OVA as the coating agent the antiserum indicated a rather

high cross reactivity value of 12.5% for G418, with I50 of 400 ng mL-1

(Figure 4.27,

Table 4.11). These values can be attributed to the high number of similar epitopes G418

shares with NB82, as can be seen in Figure 4.27.

Cross reactivity values of the structurally related synthetic derivatives NB74,

NB84, NB124 and NB125 where shown to range from 0.4-4%, with I50 values of 1.25-12

μg mL-1

(Table 4.11). These compounds are derived from G418; therefore they all

contain the NB82 scaffold. The observed 1- to 2-orders of magnitude gaps in cross

reactivity values (when compared to G418) can be explained by the difference in their

substitution pattern regarding the 2-deoxy-streptamine (2-DOS) ring (ring II, Figure

4.27). NB compounds are 4,5 di-substituted 2-DOS derivatives, whereas G418 is a 4,6 di-

substituted 2-DOS member. Since immunization conjugate was linked via the N-1

position, it is reasonable to hypothesize that the epitopes detected by the various antibody

populations are located in the 1st sugar ring (ring I) and the glycosidic bond relative

positions of the 2-DOS ring (ring II); as far as possible from the N-1 position. Following

this line of thought, compounds of NB-series substituted with ring III at C5 position will

generate greater steric interference with the antibody than that of G418 in which ring III

is substituted at C6 position. According to this rationale, compounds that differ only in

the substitution pattern at N-1 position should have similar affinities to the antibody.

Indeed, as can be seen from the data in Table 4.11, NB74 and NB84 that differ at their

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 104: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

92

N-1 position demonstrated similar cross reactivity values of 3.5±0.15 µg mL-1

and

3.1±0.75 µg mL-1

, respectively. This is the direct result of our design strategy of the

conjugation hapten. Cross reactivity values for compounds that did not contain NB82 as a

main scaffold were shown to be less than 0.04%, with I50 values of 130-2000 μg mL-1

(NB122, NB123, paromomycin, neomycin, gentamicin, kanamycin A and neamine,

Figure 4.31, Table 4.11).

Interestingly, the antibody populations generated were able to differentiate

between the diastereomeric compounds that differ only by the configuration at a single

chiral carbon center. Among the series of compounds tested in this study we had two

such pairs of compounds: NB122 (5"-S) vs NB123 (5"-R), and NB124 (5"-S) vs NB125

(5"-R). The I50 values of NB125 (5"-R) were 1 order of magnitude lower than those

obtained for its 5"-diastereomer, NB124 (5"-S) (Table 4.11). Similarly, the values

obtained for NB123 (5"-R) were twice lower than those obtained for NB122 (5"-S).

These results demonstrate a stereo-selective preference towards the 5"-R isomers. An

opposite tendency was observed for these compounds regarding their readthtough activity

and cytotoxicity values 33, 34, 82, 83, 85, 87-91

. These observations could be attributed to the

different pattern of interactions between the 5''-groups and the antibody binding pocket.

Figure 4.31: Homologous assay cross reactivity. Antiserum cross reactivity of natural and synthetic

representative AG derivatives: NB82 (♦), geneticin (G418) (●), NB125 (◊), paromamine (□), NB84 (■),

NB74 (▲), NB124 ( ), gentamicin (○), NB123 (■), paromomycin (+), NB122 (▲), neomycin (-),

kanamycin A (X), and neamine ( ). NB82-OVA was used as coating antigen (dilution 1:8,000); Antiserum

dilution (Rabbit 8245) 1:80,000. Each value represents the mean of at least 3 experiments performed in

duplicates. Mean I50 values ±S.E.M. are summarized in Table 4.11.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 105: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

93

4.2.3 Development of Heterologous-Assays

The cross reactivity values obtained in the homologous assays demonstrated the

antibodies ability to bind a wide range of synthetic and natural derivatives. However, the

enhanced affinity of the antibodies towards the homologous coating conjugate (NB82-

OVA) used in these assays resulted in a 1-5 orders of magnitude gap between the

affinities to NB82 and the other tested compounds. We assumed that in the homologous

ELISA used, the specific antibodies, induced by the NB82-KLH immunogen conjugate,

might bind the homologous coating conjugate NB82-OVA with such a high affinity that

competition with the free hapten (NB82) or other similar structures will only be visible at

high solution concentrations of the free antigen. Previous studies have demonstrated that

using heterologous conjugates as coating antigens can significantly enhance the

sensitivity of such assays towards different antigens 158

. A heterologous ELISA refers to a

system in which the haptens used for the immunogen and for the coating conjugate have

slightly different chemical structures 158

. In this case, a heterologous coating conjugate

will have a lower affinity to the antibody, and consequently, the free hapten and or

similar structures will be able to displace the antibodies more sensitively. Therefore, in

attempts to enhance the sensitivity of our assay, we developed heterologus ELISAs

towards 2 leading NB compounds, NB84 and NB124, which have shown the greatest

activity in ex-vivo and in-vivo experimentations 89, 176

. Three heterologous-coating

conjugates were prepared using the glutaraldehyde conjugation method: NB82-GTA-

OVA (control), NB84-GTA-OVA and NB124-GTA-OVA. Conjugate preparation was

followed by the development of 3 indirect heterologous-assays to test cross reactivity

values of 3 representative semi-synthetic members. The obtained results indicated a

major improvement in heterologous-antigen recognition, where I50 values for NB82,

NB84 and NB124 were reduced from 50±6 ng mL-1

to 10±3 ng mL-1

, 3.1±0.75 μg mL-1

to 0.5±0.04 μg mL-1

and 12±1 μg mL-1

to 1±0.12 μg mL-1

, respectively (Table 4.12).

Detection limits, while using the heterologous-assays were 0.001±0.0003 μg mL-1

for

NB82, 0.02±0.007 μg mL-1

for NB84 and 0.015±0.008 μg mL-1

for NB124 (Table 4.12).

The values obtained for the NB82-GTA-OVA conjugate indicated no significant

difference.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 106: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

94

Table 4.12: Concentrations at 50% inhibition (I50) and limits of detection (I20) of 3 representative

members (NB82, NB84 and NB124) using the heterologous ELISAsb.

Compound Coating antigen I50

(µg/mL) Detection limit (I20) (µg/mL)

NB82 NB82-OVA 0.05±0.006 0.008±0.001

NB82-GTA-OVA 0.05±0.009 0.007±0.001

NB84-GTA-OVA 0.04±0.006 0.007±0.001

NB124-GTA-OVA 0.01±0.003 0.001±0.0003

NB84 NB82-OVA 3.1±0.75 0.7±0.1

NB82-GTA-OVA 6±0.3 0.5±0.1

NB84-GTA-OVA 0.5±0.04 0.02±0.007

NB124-GTA-OVA 0.8±0.03 0.03±0.009

NB124 NB82-OVA 12±1.1 3±0.7

NB82-GTA-OVA 7±0.5 0.5±0.06

NB84-GTA-OVA 2±0.1 0.03±0.009

NB124-GTA-OVA 1±0.12 0.015±0.008

b NB82-OVA conjugate dilution was 1:8,000; NB82-GTA-OVA conjugate dilution was 1:128,000; NB84-GTA-OVA conjugate dilution was 1:64,000;

NB124-GTA-OVA conjugate dilution was 1:16,000; Antisera dilution (Rabbit 8245) 1:80,000. Each value represents the mean±S.E.M. of at least 3

experiments performed in duplicates.

4.2.4 Determination of Selected NB Compounds Serum Levels in Mice

Our next challenge was to use the pre-developed assay to monitor AG content in

mice serum in-vivo. For this purpose, we initially determined the antibodies affinities and

detection limits to AG in serum. We prepared 24 serial dilutions of 3 representative

members NB82, NB84 and NB124 in PBS, FBS and mice serum and quantified them by

using our pre-developed ELISA. The selection of NB84 in these experiments is based on

our recent results obtained with this designed AG as a potential treatment of

mucopolysaccharidosis type-I-Hurler (MPS I-H) disease caused by nonsense mutation 89

.

In the latter study we investigated the efficiency of a series of different suppression

therapy agents including the conventional AG gentamicin, G418, amikacin, and

paromomycin along with the designed AG NB54 and NB84, to suppress the idua-W392X

nonsense mutation in an MPS I-H mouse model. We found that NB84 suppressed the

idua-W392X nonsense mutation much more efficiently than any of the other compounds

tested. In-vivo administration of NB84 to idua-W392X mice significantly reduced urine

glycosaminoglycan (GAG) excretion and tissue GAG storage. These studies

demonstrated for the first time that NB84-mediated suppression therapy has the potential

to attenuate the MPS I-H disease phenotype. Preliminary tests of NB124, which is the

most lately developed lead compound, on the same disease models showed that it is much

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 107: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

95

better than NB84 176

. NB82 was used as a positive control to test serum interference with

ELISA protocol. The measured I50 and limit of detection (I20) values for all three

compounds tested (NB82, NB84 and NB124) in mice serum were similar to those

observed in PBS solutions (Table 4.13). Next, we determined the recovery of AG from

whole mice blood samples by spiking 250 μL blood samples with 10, 25 or 50 μg mL-1

of

the relevant compound, followed by serum extraction. Recovery ratios were determined

to be 97-102% for NB82, 92-105% for NB84 and 100-107% for NB124 (Table 4.14).

Table 4.13: Concentrations at 50% inhibition (I50) and limits of detection (I20) of 3 representative

members (NB82, NB84 and NB124) in mice serumc.

Compound Matrics I50

(µg/mL) Detection limit (I20) (µg/mL)

NB82 PBS 0.01±0.003 0.001±0.0003

Serum 0.008±0.001 0.001±0.0002

NB84 PBS 0.5±0.04 0.02±0.007

Serum 0.4±0.1 0.02±0.009

NB124 PBS 1±0.12 0.015±0.008

Serum 0.8±0.06 0.01±0.005

c For NB82 and NB124 detection: NB124-GTA-OVA conjugate dilution 1:16,000 was used as coating antigen; For NB84 detection: NB84-GTA-OVA

conjugate dilution 1:64,000 was used as coating antigen; Antisera dilution for all experiments was 1:80,000 (Rabbit 8245). Each value represents the

mean±S.E.M. of at least 3 experiments performed in duplicates.

Table 4.14: Recovery of NB82, NB84 and NB124 from whole blood samples d.

Compound Spiked concentration (µg/mL) Detected concentration (µg/mL) % Recovery

NB82 10 10.0±0.0 011

25 24.2±0.5 97

50 50.8±0.8 102

NB84 10 9.9±0.2 99

25 26.3±0.3 105

50 46.1±1.5 92

NB124 10 10.3±0.1 103

25 26.7±0.7 107

50 50.0±0.1 100

d Each value represents the mean±S.E.M. of at least 3 experiments performed in duplicates.

To determine serum levels of the above selected NB compounds in-vivo we

carried out IP injections at therapeutic doses (28.5 mg/kg body weight) to mice, and

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 108: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

96

collected blood samples at various time points post injection. Similar doses of NB84

(30.0 mg/kg body weight) were previously shown to enhance therapeutic effect in-vivo in

Idua-W392X model mice for MPS I-H disease without causing any detectable side

effects 89

. ELISA measurements of AG content in serum samples indicated that the serum

concentrations of NB84 and NB124 reached the peak levels of 121.6 μg mL-1

and 140 μg

mL-1

, respectively, within 10 min post injection, and then declined to ~10 μg mL-1

and 28

μg mL-1

by 60 min after injection (Figure 4.32). Remarkably similar kinetics were

previously reported for the AG gentamicin and amikacin detected in mice serum 167, 168

.

The obtained results also correspond to the alpha phase in the 3-compartment

pharmacokinetic model of AG serum availability, of which, a fast deterioration is

observed in the first 60 min after injection 177

.

Figure 4.32: Serum levels of NB84 and NB124 following a single dose IP injection in male mice (C57/BL,

2.5-3.5 months old), Each point represents the AG content (NB84 - black parallelogram, NB124 - white

triangle) in a single blood sample taken from different mice at various time points following the injection of

28 μg/gr body weight NB compound. The concentrations were determined using the pre-developed

heterologous-immunoassays. Each sample was prepared at 5 different concentrations and tested in

duplicates.

Due to the relatively high toxicity values observed upon AG administration,

monitoring the serum levels of these compounds in treated patients is highly important.

According to the Physicians Desk Reference (PDR) 178

, the recommended intravenous

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 109: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

97

(IV) doses of gentamicin and amikacin for antibacterial treatment in patients are 6-7.5

mg/kg/day and 15 mg/kg/day, respectively. The maximum peak serum levels allowed for

these compounds are 12 μg mL-1

and 35 μg mL-1

. However, in a recent clinical trial,

performed in France, testing the potential use of gentamicin for the treatment of CF

patients carrying various nonsense mutations, higher gentamicin peak serum levels of 20-

40 μg mL-1

were allowed 179

. These values are 2- to 4-fold higher than those approved for

antibacterial treatment. No toxic effects were reported using the declared doses during the

15 day trial, of which gentamicin was administrated once daily at 10 mg/kg body weight.

Our previous studies have indicated that all the compounds of NB-series used in this

study lack antibacterial activity and exhibit significantly lower toxicity than that of

gentamicin 33, 34, 82, 83, 85, 87-91

. However, whether the observed peak levels of 121.6 μg

mL-1

and 140 μg mL-1

, upon the administration of ~30 mg/kg body weight of NB 84 and

NB 124, are within the safety doses or not for the administration in humans, is not clear

yet and requires further investigation. Nevertheless, the sensitivity and versatility of the

generic immunoassay developed in this study are adequate for the detection of NB84 and

NB124; lowest detection limits of 20 ng mL-1

and 15 ng mL-1

, respectively that are far

below the peak levels of these compounds observed in mice serum.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 110: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

98

5. Summary and Conclusions

The present work aimed at exploring AG mechanisms of action in eukaryotes

towards the rational development of new and improved therapeutic compounds. The

structural work performed together with the obtained biochemical data enabled us to

better understand AG mechanisms of action in eukaryotic protozoa. Due to the high

similarity to human ribosomes, our Leishmania structural data also supplies indirect

evidence regarding AG action in humans. The detailed information obtained at the atomic

level also added some more benefits in our understanding of structure-activity

relationships of AG action in eukaryotes; this information is highly beneficial in the

rational design and development of new derivatives as potential therapeutic agents to

treat leishmaniasis and genetic disorders, and is currently being used in our group in

several ongoing studies.

The development of a highly sensitive methodology for the detection of the potential

therapeutic derivatives in biological samples altogether with its application for in-vivo

monitoring of selected AG in serum has also been demonstrated within the course of the

present work. This methodology is already being in an extensive use in our group and in

collaboration with additional groups working towards the clinical development of the

therapeutic compounds.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 111: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

99

Appendix

Table I Crystal screen and initial crystallization conditions determination a

Salt Precipitant Polyamine Buffer

A1 10 mM LiCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

A2 100 mM LiCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

A3 250 mM LiCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

A4 10 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

A5 100 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

A6 250 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

A7 10 mM LiCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

A8 100 mM LiCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

A9 250 mM LiCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

A10 10 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

A11 100 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

A12 250 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

B1 10 mM NaCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

B2 100 mM NaCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

B3 250 mM NaCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

B4 10 mM NaCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

B5 100 mM NaCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

B6 250 mM NaCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

B7 10 mM NaCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

B8 100 mM NaCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

B9 250 mM NaCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

B10 10 mM NaCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

B11 100 mM NaCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

B12 250 mM NaCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

C1 10 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

C2 100 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

C3 250 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

C4 10 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

C5 100 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

C6 250 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

C7 10 mM KCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

C8 100 mM KCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

C9 250 mM KCl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

C10 10 mM KCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

C11 100 mM KCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

C12 250 mM KCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

D1 10 mM NH4Cl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

D2 100 mM NH4Cl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

D3 250 mM NH4Cl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

D4 10 mM NH4Cl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 112: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

100

D5 100 mM NH4Cl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

D6 250 mM NH4Cl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

D7 10 mM NH4Cl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

D8 100 mM NH4Cl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

D9 250 mM NH4Cl 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

D10 10 mM NH4Cl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

D11 100 mM NH4Cl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

D12 250 mM NH4Cl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

E1 10 mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

E2 100 mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

E3 250 mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

E4 10 mM MgCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

E5 100 mM MgCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

E6 250 mM MgCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

E7 10 mM MgCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

E8 100 mM MgCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

E9 250 mM MgCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

E10 10 mM MgCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

E11 100 mM MgCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

E12 250 mM MgCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

F1 10 mM CaCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

F2 100 mM CaCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

F3 250 mM CaCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

F4 10 mM CaCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

F5 100 mM CaCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

F6 250 mM CaCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

F7 10 mM CaCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

F8 100 mM CaCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

F9 250 mM CaCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

F10 10 mM CaCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

F11 100 mM CaCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

F12 250 mM CaCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

G1 10 mM SrCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

G2 100 mM SrCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

G3 250 mM SrCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

G4 10 mM SrCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

G5 100 mM SrCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

G6 250 mM SrCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

G7 10 mM SrCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

G8 100 mM SrCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

G9 250 mM SrCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

G10 10 mM SrCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

G11 100 mM SrCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

G12 250 mM SrCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

H1 10 mM BaCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

H2 100 mM BaCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 113: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

101

H3 250 mM BaCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

H4 10 mM BaCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

H5 100 mM BaCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

H6 250 mM BaCl2 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 7.0)

H7 10 mM BaCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

H8 100 mM BaCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

H9 250 mM BaCl2 40% MPD 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

H10 10 mM BaCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

H11 100 mM BaCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0)

H12 250 mM BaCl2 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 7.0) a Conditions highlighted in blue were found to enhance crystal growth.

Table II Crystal optimization matrix

Salt Precipitant Polyamine Buffer

1 10 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

2 50 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

3 100 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

4 150 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

5 200 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

6 250 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

7 300 mM LiCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

8 50 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

9 100 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

10 150 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

11 200 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

12 250 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

13 300 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

14 50 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

15 100 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

16 150 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

17 200 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

18 250 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

19 50 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

20 100 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

21 150 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

22 200 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

23 250 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

24 50 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

25 100 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

26 150 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

27 200 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

28 250 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

29 300 mM KCl 20% PEG3350 1 mM Spermine 50 mM Na Cacodylate (pH 6.0/7.0)

30 50 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.0/5.5/6.0/7.0/7.5)

31 100 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.0/5.5/6.0/7.0/7.5)

32 150 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.0/5.5/6.0/7.0/7.5)

33 200 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.0/5.5/6.0/7.0/7.5)

34 250 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

35 300 mM KCl 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

36 50 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

37 100 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 114: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

102

38 150 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

39 200 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

40 250 mM KCl + 5mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

41 50 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

42 100 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

43 150 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

44 200 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

45 250 mM KCl + 10mM MgCl2 40% MPD 1 mM Spermine 50 mM Na Cacodylate (pH 5.5/6.0/7.0/7.5)

46 50 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

47 100 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

48 150 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

49 200 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

50 250 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

51 300 mM LiCl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

52 100 mM NH4Cl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

53 150 mM NH4Cl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

54 200 mM NH4Cl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

55 250 mM NH4Cl 20% PEG3350 1 mM CoHex 50 mM Na Cacodylate (pH 6.0/7.0)

a Conditions highlighted in blue were found to result in the best shaped crystals suitable for X-ray analysis.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 115: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

103

References

(1) Davis BD. Mechanism of bactericidal action of aminoglycosides. Microbiol Rev

1987;51(3):341-350.

(2) Jana S, Deb JK. Molecular understanding of aminoglycoside action and

resistance. Appl Microbiol Biotechnol 2006;70(2):140-150.

(3) Botto R, Coxon B. Nitrogen-15 nuclear magnetic resonance spectroscopy of

neomycin B and related aminoglycosides. J Med Chem 1993;105:1021-1028.

(4) Kaul M, Barbieri CM, Kerrigan JE, Pilch DS. Coupling of drug protonation to

the specific binding of aminoglycosides to the A site of 16 S rRNA: elucidation

of the number of drug amino groups involved and their identities. J Mol Biol

2003;326(5):1373-1387.

(5) von AU, Davies J, Schroeder R. Antibiotic inhibition of group I ribozyme

function. Nature 1991;353(6342):368-370.

(6) Waldsich C, Semrad K, Schroeder R. Neomycin B inhibits splicing of the td

intron indirectly by interfering with translation and enhances missplicing in

vivo. RNA 1998;4(12):1653-1663.

(7) Mei HY, Cui M, Heldsinger A et al. Inhibitors of protein-RNA complexation

that target the RNA: specific recognition of human immunodeficiency virus

type 1 TAR RNA by small organic molecules. Biochemistry

1998;37(40):14204-14212.

(8) Zapp ML, Stern S, Green MR. Small molecules that selectively block RNA

binding of HIV-1 Rev protein inhibit Rev function and viral production. Cell

1993;74(6):969-978.

(9) Singh N, Kumar M, Singh RK. Leishmaniasis: Current status of available drugs

and new potential drug targets. Asian Pacific Journal of Tropical Medicine

2012;5(6):485-497.

(10) Bidou L, Allamand V, Rousset JP, Namy O. Sense from nonsense: therapies for

premature stop codon diseases. Trends in Molecular Medicine 2012;18(11):679-

688.

(11) Hainrichson M, Nudelman I, Baasov T. Designer aminoglycosides: the race to

develop improved antibiotics and compounds for the treatment of human

genetic diseases. Organic & Biomolecular Chemistry 2008;6(2):227-239.

(12) Keeling KM, Wang D, Conard SE, Bedwell DM. Suppression of premature

termination codons as a therapeutic approach. Critical Reviews in Biochemistry

and Molecular Biology 2012;47(5):444-463.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 116: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

104

(13) Moazed D, Noller HF. Interaction of antibiotics with functional sites in 16S

ribosomal RNA. Nature 1987;327(6121):389-394.

(14) Moazed D, Noller HF. Binding of tRNA to the ribosomal A and P sites protects

two distinct sets of nucleotides in 16 S rRNA. J Mol Biol 1990;211(1):135-145.

(15) Mueller F, Stark H, van HM, Rinke-Appel J, Brimacombe R. A new model for

the three-dimensional folding of Escherichia coli 16 S ribosomal RNA. III. The

topography of the functional centre. J Mol Biol 1997;271(4):566-587.

(16) O'Connor M, Brunelli CA, Firpo MA et al. Genetic probes of ribosomal RNA

function. Biochem Cell Biol 1995;73(11-12):859-868.

(17) Ogle JM, Brodersen DE, Clemons WM, Jr., Tarry MJ, Carter AP,

Ramakrishnan V. Recognition of cognate transfer RNA by the 30S ribosomal

subunit. Science 2001;292(5518):897-902.

(18) Jenner L, Demeshkina N, Yusupova G, Yusupov M. Structural rearrangements

of the ribosome at the tRNA proofreading step. Nat Struct Mol Biol

2010;17(9):1072-1078.

(19) Kondo J, Westhof E. The bacterial and mitochondrial ribosomal A-site

molecular switches possess different conformational substates. Nucleic Acids

Res 2008;36(8):2654-2666.

(20) Gorini L, Kataja E. Phenotypic Repair By Streptomycin Of Defective

Genotypes In E. coli. Proc Natl Acad Sci U S A 1964;51:487-493.

(21) OMIM. 2011.

(22) Atkinson J, Martin R. Mutations to nonsense codons in human genetic disease:

implications for gene therapy by nonsense suppressor tRNAs. Nucleic Acids Res

1994;22(8):1327-1334.

(23) Kellermayer R. Translational readthrough induction of pathogenic nonsense

mutations. Eur J Med Genet 2006;49(6):445-450.

(24) Burke JF, Mogg AE. Suppression of a nonsense mutation in mammalian cells in

vivo by the aminoglycoside antibiotics G-418 and paromomycin. Nucleic Acids

Res 1985;13(17):6265-6272.

(25) Kaufman RJ. Correction of genetic disease by making sense from nonsense. J

Clin Invest 1999;104(4):367-368.

(26) Manuvakhova M, Keeling K, Bedwell DM. Aminoglycoside antibiotics mediate

context-dependent suppression of termination codons in a mammalian

translation system. RNA 2000;6(7):1044-1055.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 117: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

105

(27) Wilschanski M, Famini C, Blau H et al. A pilot study of the effect of gentamicin

on nasal potential difference measurements in cystic fibrosis patients carrying

stop mutations. Am J Respir Crit Care Med 2000;161(3 Pt 1):860-865.

(28) Wilschanski M, Yahav Y, Yaacov Y et al. Gentamicin-induced correction of

CFTR function in patients with cystic fibrosis and CFTR stop mutations. N Engl

J Med 2003;349(15):1433-1441.

(29) Malik V, Rodino-Klapac LR, Viollet L, Mendell JR. Aminoglycoside-induced

mutation suppression (stop codon readthrough) as a therapeutic strategy for

Duchenne muscular dystrophy. Ther Adv Neurol Disord 2010;3(6):379-389.

(30) Barton-Davis ER, Cordier L, Shoturma DI, Leland SE, Sweeney HL.

Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of

mdx mice. J Clin Invest 1999;104(4):375-381.

(31) Malik V, Rodino-Klapac LR, Viollet L et al. Gentamicin-induced readthrough

of stop codons in Duchenne muscular dystrophy. Ann Neurol 2010;67(6):771-

780.

(32) Kerem E. Pharmacologic therapy for stop mutations: how much CFTR activity

is enough? Curr Opin Pulm Med 2004;10(6):547-552.

(33) Brendel C, Belakhov V, Werner H et al. Readthrough of nonsense mutations in

Rett syndrome: evaluation of novel aminoglycosides and generation of a new

mouse model. J Mol Med (Berl) 2011;89(4):389-398.

(34) Vecsler M, Ben ZB, Nudelman I et al. Ex vivo treatment with a novel synthetic

aminoglycoside NB54 in primary fibroblasts from Rett syndrome patients

suppresses MECP2 nonsense mutations. PLoS One 2011;6(6):e20733.

(35) Keeling KM, Brooks DA, Hopwood JJ, Li P, Thompson JN, Bedwell DM.

Gentamicin-mediated suppression of Hurler syndrome stop mutations restores a

low level of alpha-L-iduronidase activity and reduces lysosomal

glycosaminoglycan accumulation. Hum Mol Genet 2001;10(3):291-299.

(36) Sangkuhl K, Schulz A, Rompler H, Yun J, Wess J, Schoneberg T.

Aminoglycoside-mediated rescue of a disease-causing nonsense mutation in the

V2 vasopressin receptor gene in vitro and in vivo. Hum Mol Genet

2004;13(9):893-903.

(37) Helip-Wooley A, Park MA, Lemons RM, Thoene JG. Expression of CTNS

alleles: subcellular localization and aminoglycoside correction in vitro. Mol

Genet Metab 2002;75(2):128-133.

(38) Grayson C, Bartolini F, Chapple JP et al. Localization in the human retina of the

X-linked retinitis pigmentosa protein RP2, its homologue cofactor C and the

RP2 interacting protein Arl3. Hum Mol Genet 2002;11(24):3065-3074.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 118: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

106

(39) Xi B, Guan F, Lawrence DS. Enhanced production of functional proteins from

defective genes. J Am Chem Soc 2004;126(18):5660-5661.

(40) Wolstencroft EC, Mattis V, Bajer AA, Young PJ, Lorson CL. A non-sequence-

specific requirement for SMN protein activity: the role of aminoglycosides in

inducing elevated SMN protein levels. Hum Mol Genet 2005;14(9):1199-1210.

(41) Floquet C, Rousset JP, Bidou L. Readthrough of premature termination codons

in the adenomatous polyposis coli gene restores its biological activity in human

cancer cells. PLoS One 2011;6(8):e24125.

(42) Floquet C, Deforges J, Rousset JP, Bidou L. Rescue of non-sense mutated p53

tumor suppressor gene by aminoglycosides. Nucleic Acids Res

2011;39(8):3350-3362.

(43) Zilberberg A, Lahav L, Rosin-Arbesfeld R. Restoration of APC gene function in

colorectal cancer cells by aminoglycoside- and macrolide-induced read-through

of premature termination codons. Gut 2010;59(4):496-507.

(44) Floquet C, Hatin I, Rousset JP, Bidou L. Statistical analysis of readthrough

levels for nonsense mutations in mammalian cells reveals a major determinant

of response to gentamicin. PLoS Genet 2012;8(3):e1002608.

(45) Howard MT, Shirts BH, Petros LM, Flanigan KM, Gesteland RF, Atkins JF.

Sequence specificity of aminoglycoside-induced stop condon readthrough:

potential implications for treatment of Duchenne muscular dystrophy. Ann

Neurol 2000;48(2):164-169.

(46) Howard MT, Anderson CB, Fass U et al. Readthrough of dystrophin stop codon

mutations induced by aminoglycosides. Ann Neurol 2004;55(3):422-426.

(47) Pfister P, Hobbie S, Vicens Q, Bottger EC, Westhof E. The molecular basis for

A-site mutations conferring aminoglycoside resistance: relationship between

ribosomal susceptibility and X-ray crystal structures. Chembiochem

2003;4(10):1078-1088.

(48) Recht MI, Douthwaite S, Puglisi JD. Basis for prokaryotic specificity of action

of aminoglycoside antibiotics. EMBO J 1999;18(11):3133-3138.

(49) Keeling KM, Bedwell DM. Suppression of nonsense mutations as a therapeutic

approach to treat genetic diseases. Wiley Interdiscip Rev RNA 2011;2(6):837-

852.

(50) Korostelev A, Zhu J, Asahara H, Noller HF. Recognition of the amber UAG

stop codon by release factor RF1. EMBO J 2010;29(15):2577-2585.

(51) Laurberg M, Asahara H, Korostelev A, Zhu J, Trakhanov S, Noller HF.

Structural basis for translation termination on the 70S ribosome. Nature

2008;454(7206):852-857.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 119: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

107

(52) Jin H, Kelley AC, Loakes D, Ramakrishnan V. Structure of the 70S ribosome

bound to release factor 2 and a substrate analog provides insights into catalysis

of peptide release. Proc Natl Acad Sci U S A 2010;107(19):8593-8598.

(53) Petry S, Brodersen DE, Murphy FV et al. Crystal structures of the ribosome in

complex with release factors RF1 and RF2 bound to a cognate stop codon. Cell

2005;123(7):1255-1266.

(54) Weixlbaumer A, Jin H, Neubauer C et al. Insights into translational termination

from the structure of RF2 bound to the ribosome. Science 2008;322(5903):953-

956.

(55) Brown CM, McCaughan KK, Tate WP. Two regions of the Escherichia coli 16S

ribosomal RNA are important for decoding stop signals in polypeptide chain

termination. Nucleic Acids Res 1993;21(9):2109-2115.

(56) Youngman EM, He SL, Nikstad LJ, Green R. Stop codon recognition by release

factors induces structural rearrangement of the ribosomal decoding center that is

productive for peptide release. Mol Cell 2007;28(4):533-543.

(57) Edlind TD. Susceptibility of Giardia lamblia to aminoglycoside protein

synthesis inhibitors: correlation with rRNA structure. Antimicrob Agents

Chemother 1989;33(4):484-488.

(58) Hobbie SN, Kaiser M, Schmidt S et al. Genetic reconstruction of protozoan

rRNA decoding sites provides a rationale for paromomycin activity against

Leishmania and Trypanosoma. PLoS Negl Trop Dis 2011;5(5):e1161.

(59) Kondo J, Koganei M, Maianti JP, Ly VL, Hanessian S. Crystal Structures of a

Bioactive 6'-Hydroxy Variant of Sisomicin Bound to the Bacterial and

Protozoal Ribosomal Decoding Sites. ChemMedChem 2013.

(60) Loebenberg D, Counels M, Waitz JA. Antibiotic G418, a new

micrommomospora-produced aminglycoside with activity apainst protozoa and

helminths: antiparasitic activity. Antimicrob Agents Chemother 1975;7(6):811-

815.

(61) Singh N, Kumar M, Singh RK. Leishmaniasis: current status of available drugs

and new potential drug targets. Asian Pac J Trop Med 2012;5(6):485-497.

(62) Davidson RN, den BM, Ritmeijer K. Paromomycin. Trans R Soc Trop Med Hyg

2009;103(7):653-660.

(63) Sinha PK, Jha TK, Thakur CP et al. Phase 4 pharmacovigilance trial of

paromomycin injection for the treatment of visceral leishmaniasis in India. J

Trop Med 2011;2011:645203.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 120: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

108

(64) Nagai J, Takano M. Molecular aspects of renal handling of aminoglycosides and

strategies for preventing the nephrotoxicity. Drug Metab Pharmacokinet

2004;19(3):159-170.

(65) Bottger EC. Antimicrobial agents targeting the ribosome: the issue of selectivity

and toxicity - lessons to be learned. Cell Mol Life Sci 2007;64(7-8):791-795.

(66) Hobbie SN, Akshay S, Kalapala SK, Bruell CM, Shcherbakov D, Bottger EC.

Genetic analysis of interactions with eukaryotic rRNA identify the

mitoribosome as target in aminoglycoside ototoxicity. Proc Natl Acad Sci U S A

2008;105(52):20888-20893.

(67) Kharkheli E, Kevanishvili Z, Maglakelidze T, Davitashvili O, Schacht J. Does

vitamin E prevent gentamicin-induced ototoxicity? Georgian Med News

2007;(146):14-17.

(68) Li G, Sha SH, Zotova E, Arezzo J, Van de Water T, Schacht J. Salicylate

protects hearing and kidney function from cisplatin toxicity without

compromising its oncolytic action. Lab Invest 2002;82(5):585-596.

(69) Sha SH, Schacht J. Salicylate attenuates gentamicin-induced ototoxicity. Lab

Invest 1999;79(7):807-813.

(70) Sha SH, Schacht J. Antioxidants attenuate gentamicin-induced free radical

formation in vitro and ototoxicity in vivo: D-methionine is a potential

protectant. Hear Res 2000;142(1-2):34-40.

(71) Sha SH, Qiu JH, Schacht J. Aspirin to prevent gentamicin-induced hearing loss.

N Engl J Med 2006;354(17):1856-1857.

(72) Song BB, Sha SH, Schacht J. Iron chelators protect from aminoglycoside-

induced cochleo- and vestibulo-toxicity. Free Radic Biol Med 1998;25(2):189-

195.

(73) Beauchamp D, Laurent G, Maldague P, Abid S, Kishore BK, Tulkens PM.

Protection against gentamicin-induced early renal alterations (phospholipidosis

and increased DNA synthesis) by coadministration of poly-L-aspartic acid. J

Pharmacol Exp Ther 1990;255(2):858-866.

(74) Gilbert DN, Wood CA, Kohlhepp SJ et al. Polyaspartic acid prevents

experimental aminoglycoside nephrotoxicity. J Infect Dis 1989;159(5):945-953.

(75) Thibault N, Grenier L, Simard M, Bergeron MG, Beauchamp D. Attenuation by

daptomycin of gentamicin-induced experimental nephrotoxicity. Antimicrob

Agents Chemother 1994;38(5):1027-1035.

(76) Thibault N, Grenier L, Simard M, Bergeron MG, Beauchamp D. Protection

against gentamicin nephrotoxicity by daptomycin in nephrectomized rats. Life

Sci 1995;56(22):1877-1887.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 121: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

109

(77) Mattis VB, Fosso MY, Chang CW, Lorson CL. Subcutaneous administration of

TC007 reduces disease severity in an animal model of SMA. BMC Neurosci

2009;10:142.

(78) Brichta L, Hofmann Y, Hahnen E et al. Valproic acid increases the SMN2

protein level: a well-known drug as a potential therapy for spinal muscular

atrophy. Hum Mol Genet 2003;12(19):2481-2489.

(79) Sumner CJ, Huynh TN, Markowitz JA et al. Valproic acid increases SMN levels

in spinal muscular atrophy patient cells. Ann Neurol 2003;54(5):647-654.

(80) Mattis VB, Ebert AD, Fosso MY, Chang CW, Lorson CL. Delivery of a read-

through inducing compound, TC007, lessens the severity of a spinal muscular

atrophy animal model. Hum Mol Genet 2009;18(20):3906-3913.

(81) Chang CW, Hui Y, Elchert B, Wang J, Li J, Rai R. Pyranmycins, a novel class

of aminoglycosides with improved acid stability: the SAR of D-pyranoses on

ring III of pyranmycin. Org Lett 2002;4(26):4603-4606.

(82) Nudelman I, Rebibo-Sabbah A, Shellom-Shezifi D et al. Redesign of

aminoglycosides for treatment of human genetic diseases caused by premature

stop mutations. Bioorg Med Chem Lett 2006;16(24):6310-6315.

(83) Rebibo-Sabbah A, Nudelman I, Ahmed ZM, Baasov T, Ben-Yosef T. In vitro

and ex vivo suppression by aminoglycosides of PCDH15 nonsense mutations

underlying type 1 Usher syndrome. Hum Genet 2007;122(3-4):373-381.

(84) Goldmann T, Rebibo-Sabbah A, Overlack N et al. Beneficial read-through of a

USH1C nonsense mutation by designed aminoglycoside NB30 in the retina.

Invest Ophthalmol Vis Sci 2010;51(12):6671-6680.

(85) Nudelman I, Rebibo-Sabbah A, Cherniavsky M et al. Development of novel

aminoglycoside (NB54) with reduced toxicity and enhanced suppression of

disease-causing premature stop mutations. J Med Chem 2009;52(9):2836-2845.

(86) Kondo S, Hotta K. Semisynthetic aminoglycoside antibiotics: Development and

enzymatic modifications. J Infect Chemother 1999;5(1):1-9.

(87) Rowe SM, Sloane P, Tang LP et al. Suppression of CFTR premature

termination codons and rescue of CFTR protein and function by the synthetic

aminoglycoside NB54. J Mol Med (Berl) 2011;89(11):1149-1161.

(88) Nudelman I, Glikin D, Smolkin B, Hainrichson M, Belakhov V, Baasov T.

Repairing faulty genes by aminoglycosides: development of new derivatives of

geneticin (G418) with enhanced suppression of diseases-causing nonsense

mutations. Bioorg Med Chem 2010;18(11):3735-3746.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 122: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

110

(89) Wang D, Belakhov V, Kandasamy J et al. The designer aminoglycoside NB84

significantly reduces glycosaminoglycan accumulation associated with MPS I-H

in the Idua-W392X mouse. Mol Genet Metab 2011.

(90) Kandasamy J, Atia-Glikin D, Belakhov V, Baasov T. Repairing faulty genes by

aminoglycosides: Identification of new pharmacophore with enhanced

suppression of disease-causing nonsense mutations . Med Chem Comm

2011;2:165-171.

(91) Kandasamy J, Atia-Glikin D, Shulman E et al. Increased selectivity toward

cytoplasmic versus mitochondrial ribosome confers improved efficiency of

synthetic aminoglycosides in fixing damaged genes: a strategy for treatment of

genetic diseases caused by nonsense mutations. J Med Chem

2012;55(23):10630-10643.

(92) Kondo J, Hainrichson M, Nudelman I et al. Differential selectivity of natural

and synthetic aminoglycosides towards the eukaryotic and prokaryotic decoding

A sites. Chembiochem 2007;8(14):1700-1709.

(93) Korostelev A, Ermolenko DN, Noller HF. Structural dynamics of the ribosome.

Curr Opin Chem Biol 2008;12(6):674-683.

(94) Steitz TA. A structural understanding of the dynamic ribosome machine. Nat

Rev Mol Cell Biol 2008;9(3):242-253.

(95) Yonath A, Bashan A. Ribosomal crystallography: initiation, peptide bond

formation, and amino acid polymerization are hampered by antibiotics. Annu

Rev Microbiol 2004;58:233-251.

(96) Schmeing TM, Ramakrishnan V. What recent ribosome structures have revealed

about the mechanism of translation. Nature 2009;461(7268):1234-1242.

(97) Frank J. Electron microscopy of functional ribosome complexes. Biopolymers

2003;68(2):223-233.

(98) Stark H. Three-dimensional electron cryomicroscopy of ribosomes. Curr

Protein Pept Sci 2002;3(1):79-91.

(99) Ben-Shem A, Jenner L, Yusupova G, Yusupov M. Crystal structure of the

eukaryotic ribosome. Science 2010;330(6008):1203-1209.

(100) Ben-Shem A, Garreau de LN, Melnikov S, Jenner L, Yusupova G, Yusupov M.

The structure of the eukaryotic ribosome at 3.0 A resolution. Science

2011;334(6062):1524-1529.

(101) Klinge S, Voigts-Hoffmann F, Leibundgut M, Arpagaus S, Ban N. Crystal

structure of the eukaryotic 60S ribosomal subunit in complex with initiation

factor 6. Science 2011;334(6058):941-948.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 123: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

111

(102) Rabl J, Leibundgut M, Ataide SF, Haag A, Ban N. Crystal structure of the

eukaryotic 40S ribosomal subunit in complex with initiation factor 1. Science

2011;331(6018):730-736.

(103) Kondo J, Francois B, Urzhumtsev A, Westhof E. Crystallographic studies of

Homo sapiens ribosomal decoding A site complexed with aminoglycosides.

Nucleic Acids Symp Ser (Oxf) 2005;(49):253-254.

(104) Hermann T, Tereshko V, Skripkin E, Patel DJ. Apramycin recognition by the

human ribosomal decoding site. Blood Cells Mol Dis 2007;38(3):193-198.

(105) Lynch SR, Puglisi JD. Structure of a eukaryotic decoding region A-site RNA. J

Mol Biol 2001;306(5):1023-1035.

(106) Tsai A, Uemura S, Johansson M et al. The impact of aminoglycosides on the

dynamics of translation elongation. Cell Rep 2013;3(2):497-508.

(107) Kaul M, Barbieri CM, Pilch DS. Defining the basis for the specificity of

aminoglycoside-rRNA recognition: a comparative study of drug binding to the

A sites of Escherichia coli and human rRNA. J Mol Biol 2005;346(1):119-134.

(108) Xie Y, Dix AV, Tor Y. Antibiotic selectivity for prokaryotic vs. eukaryotic

decoding sites. Chem Commun (Camb ) 2010;46(30):5542-5544.

(109) Kythreoti G, Vourloumis D. A Homo sapiens cytoplasmic ribosomal decoding

A-site affinity screen evaluating aminoglycoside and analogue binding. Anal

Biochem 2011;412(1):102-107.

(110) Francois B, Russell RJ, Murray JB et al. Crystal structures of complexes

between aminoglycosides and decoding A site oligonucleotides: role of the

number of rings and positive charges in the specific binding leading to

miscoding. Nucleic Acids Res 2005;33(17):5677-5690.

(111) Hobbie SN, Pfister P, Brull C, Westhof E, Bottger EC. Analysis of the

contribution of individual substituents in 4,6-aminoglycoside-ribosome

interaction. Antimicrob Agents Chemother 2005;49(12):5112-5118.

(112) Kondo J, Pachamuthu K, Francois B, Szychowski J, Hanessian S, Westhof E.

Crystal structure of the bacterial ribosomal decoding site complexed with a

synthetic doubly functionalized paromomycin derivative: a new specific binding

mode to an a-minor motif enhances in vitro antibacterial activity.

ChemMedChem 2007;2(11):1631-1638.

(113) Vicens Q, Westhof E. Crystal structure of paromomycin docked into the

eubacterial ribosomal decoding A site. Structure 2001;9(8):647-658.

(114) Vicens Q, Westhof E. Crystal structure of geneticin bound to a bacterial 16S

ribosomal RNA A site oligonucleotide. J Mol Biol 2003;326(4):1175-1188.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 124: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

112

(115) Francis SP, Katz J, Fanning KD et al. A novel role of cytosolic protein synthesis

inhibition in aminoglycoside ototoxicity. J Neurosci 2013;33(7):3079-3093.

(116) Scheunemann AE, Graham WD, Vendeix FA, Agris PF. Binding of

aminoglycoside antibiotics to helix 69 of 23S rRNA. Nucleic Acids Res

2010;38(9):3094-3105.

(117) Hainrichson M, Nudelman I, Baasov T. Designer aminoglycosides: the race to

develop improved antibiotics and compounds for the treatment of human

genetic diseases. Org Biomol Chem 2008;6(2):227-239.

(118) Kondo J, Francois B, Urzhumtsev A, Westhof E. Crystal structure of the Homo

sapiens cytoplasmic ribosomal decoding site complexed with apramycin. Angew

Chem Int Ed Engl 2006;45(20):3310-3314.

(119) Lynch SR, Puglisi JD. Structural origins of aminoglycoside specificity for

prokaryotic ribosomes. J Mol Biol 2001;306(5):1037-1058.

(120) Fourmy D, Recht MI, Blanchard SC, Puglisi JD. Structure of the A site of

Escherichia coli 16S ribosomal RNA complexed with an aminoglycoside

antibiotic. Science 1996;274(5291):1367-1371.

(121) Kondo J, Urzhumtsev A, Westhof E. Two conformational states in the crystal

structure of the Homo sapiens cytoplasmic ribosomal decoding A site. Nucleic

Acids Res 2006;34(2):676-685.

(122) Kabsch W. XDS. Acta Crystallogr D Biol Crystallogr 2010;66(Pt 2):125-132.

(123) Battye TG, Kontogiannis L, Johnson O, Powell HR, Leslie AG. iMOSFLM: a

new graphical interface for diffraction-image processing with MOSFLM. Acta

Crystallogr D Biol Crystallogr 2011;67(Pt 4):271-281.

(124) The CCP4 suite: programs for protein crystallography. Acta Crystallogr D Biol

Crystallogr 1994;50(Pt 5):760-763.

(125) Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta

Crystallogr D Biol Crystallogr 2004;60(Pt 12 Pt 1):2126-2132.

(126) Adams PD, Afonine PV, Bunkoczi G et al. PHENIX: a comprehensive Python-

based system for macromolecular structure solution. Acta Crystallogr D Biol

Crystallogr 2010;66(Pt 2):213-221.

(127) Brunger AT, Adams PD, Clore GM et al. Crystallography & NMR system: A

new software suite for macromolecular structure determination. Acta

Crystallogr D Biol Crystallogr 1998;54(Pt 5):905-921.

(128) Brunger AT. Version 1.2 of the Crystallography and NMR system. Nat Protoc

2007;2(11):2728-2733.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 125: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

113

(129) The PyMOL Molecular Graphics System V1504SL. The PyMOL Molecular

Graphics System, Version 1.5.0.4 Schrצdinger, LLC.

Ref Type: Generic

(130) Bergfors T. Screening and optimization methods for nonautomated

crystallization laboratories. Methods Mol Biol 2007;363:131-151.

(131) Shimony O, Jaffe CL. Rapid fluorescent assay for screening drugs on

Leishmania amastigotes. J Microbiol Methods 2008;75(2):196-200.

(132) Chen Y, Shang Y, Li X, Wu X, Xiao X. Development of an enzyme-linked

immunoassay for the detection of gentamicin in swine tissues. Food Chemistry

2008;108:304-309.

(133) Kondo J, Hainrichson M, Nudelman I et al. Differential selectivity of natural

and synthetic aminoglycosides towards the eukaryotic and prokaryotic decoding

A sites. Chembiochem 2007;8(14):1700-1709.

(134) Ashe MP, De Long SK, Sachs AB. Glucose depletion rapidly inhibits

translation initiation in yeast. Molecular Biology of the Cell 2000;11(3):833-

848.

(135) Balagopal V, Parker R. Stm1 modulates translation after 80S formation in

Saccharomyces cerevisiae. Rna-A Publication of the Rna Society

2011;17(5):835-842.

(136) Kondo J. A Structural Basis for the Antibiotic Resistance Conferred by an

A1408G Mutation in 16S rRNA and for the Antiprotozoal Activity of

Aminoglycosides. Angew Chem Int Ed Engl 2011;50:1-5.

(137) Kondo J. A structural basis for the antibiotic resistance conferred by an A1408G

mutation in 16S rRNA and for the antiprotozoal activity of aminoglycosides.

Angew Chem Int Ed Engl 2012;51(2):465-468.

(138) Fernandez MM, Malchiodi EL, Algranati ID. Differential effects of

paromomycin on ribosomes of Leishmania mexicana and mammalian cells.

Antimicrob Agents Chemother 2011;55(1):86-93.

(139) Matt T, Ng CL, Lang K et al. Dissociation of antibacterial activity and

aminoglycoside ototoxicity in the 4-monosubstituted 2-deoxystreptamine

apramycin. Proceedings of the National Academy of Sciences of the United

States of America 2012;109(27):10984-10989.

(140) Pfister P, Hobbie S, Brull C et al. Mutagenesis of 16S rRNA C1409-G1491

base-pair differentiates between 6'OH and 6'NH3+ aminoglycosides. J Mol Biol

2005;346(2):467-475.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 126: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

114

(141) Jhingran A, Chawla B, Saxena S, Barrett MP, Madhubala R. Paromomycin:

uptake and resistance in Leishmania donovani. Mol Biochem Parasitol

2009;164(2):111-117.

(142) Zufferey R, Bibis SS, Zhu T, Dhalladoo S. Characterization of a compensatory

mutant of Leishmania major that lacks ether lipids but exhibits normal growth,

and G418 and hygromycin resistance. Exp Parasitol 2012;130(3):200-204.

(143) Pokrovskaya V, Belakhov V, Hainrichson M, Yaron S, Baasov T. Design,

Synthesis, and Evaluation of Novel Fluoroquinolone-Aminoglycoside Hybrid

Antibiotics. Journal of Medicinal Chemistry 2009;52(8):2243-2254.

(144) Han Q, Zhao Q, Fish S et al. Molecular recognition by glycoside pseudo base

pairs and triples in an apramycin-RNA complex. Angew Chem Int Ed Engl

2005;44(18):2694-2700.

(145) Haasnoot W, Stouten P, Cazemier G, Lommen A, Nouws JF, Keukens HJ.

Immunochemical detection of aminoglycosides in milk and kidney. Analyst

1999;124(3):301-305.

(146) Kampsholtzapple C, Stanker LH, Deloach JR. Development of A Monoclonal

Antibody-Based Elisa for the Anthelmintic Hygromycin-B. Journal of

Agricultural and Food Chemistry 1994;42(3):822-827.

(147) Solomun B, Bilandzic N, Varenina I, Scortichini G. Validation of an enzyme-

linked immunosorbent assay for qualitative screening of neomycin in muscle,

liver, kidney, eggs and milk. Food Addit Contam Part A Chem Anal Control

Expo Risk Assess 2011;28(1):11-18.

(148) Abuknesha RA, Luk C. Enzyme immunoassays for the analysis of streptomycin

in milk, serum and water: development and assessment of a polyclonal

antiserum and assay procedures using novel streptomycin derivatives. Analyst

2005;130(6):964-970.

(149) Jin Y, Jang JW, Lee MH, Han CH. Development of competitive direct enzyme-

linked immunosorbent assay for the detection of gentamicin residues in the

plasma of live animals. Asian-Australasian Journal of Animal Sciences

2005;18(10):1498-1504.

(150) Jin Y, Jang JW, Han CH, Lee MH. Development of ELISA and

immunochromatographic assay for the detection of gentamicin. Journal of

Agricultural and Food Chemistry 2005;53(20):7639-7643.

(151) Jin Y, Jang JW, Han CH, Lee MH. Development of immunoassays for the

detection of kanamycin in veterinary fields. Journal of Veterinary Science

2006;7(2):111-117.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 127: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

115

(152) Jin Y, Jang JW, Lee MH, Han CH. Development of ELISA and

immunochromatographic assay for the detection of neomycin. Clinica Chimica

Acta 2006;364(1-2):260-266.

(153) Kolosova AY, Blintsov AN, Samsonova JV, Egorov AM. Development of an

enzyme-linked immunosorbent assay for gentamicin in human blood serum.

Fresenius Journal of Analytical Chemistry 1998;361(3):329-330.

(154) Watanabe H, Satake A, Kido Y, Tsuji A. Production of monoclonal antibody

and development of enzyme-linked immunosorbent assay for kanamycin in

biological matrices. Analyst 1999;124(11):1611-1615.

(155) Ara J, Gans Z, Sweeney R, Wolf B. Dot-Elisa for the Rapid Detection of

Gentamicin in Milk. Journal of Clinical Laboratory Analysis 1995;9(5):320-

324.

(156) Byzova NA, Zvereva EA, Zherdev AV, Eremin SA, Sveshnikov PG, Dzantiev

BB. Pretreatment-free immunochromatographic assay for the detection of

streptomycin and its application to the control of milk and dairy products.

Analytica Chimica Acta 2011;701(2):209-217.

(157) Galvidis IA, Burkin MA. Monoclonal antibody-based enzyme-linked

immunosorbent assay for the aminoglycoside antibiotic kanamycin in

foodstuffs. Russian Journal of Bioorganic Chemistry 2010;36(6):722-729.

(158) Loomans EE, Van WJ, Koets M, Van AA. Neamin as an immunogen for the

development of a generic ELISA detecting gentamicin, kanamycin, and

neomycin in milk. J Agric Food Chem 2003;51(3):587-593.

(159) Samsonova JV, Bashkurov ML, Ivanova NL, Rubtsova MY, Egorov AM.

ELISA of streptomycin in buffer and milk: Effect of reagents' structure and

analysis format on assay performance. Food and Agricultural Immunology

2005;16(1):47-57.

(160) Xu NF, Qu CL, Ma W et al. Development and application of one-step ELISA

for the detection of neomycin in milk. Food and Agricultural Immunology

2011;22(3):259-269.

(161) Preu M, Guyot D, Petz M. Development of a gas chromatography mass

spectrometry method for the analysis of aminoglycoside antibiotics using

experimental design for the optimisation of the derivatisation reactions. Journal

of Chromatography A 1998;818(1):95-108.

(162) Hubicka U, Krzek J, Woltynska H, Stachacz B. Simultaneous Identification and

Quantitative Determination of Selected Aminoglycoside Antibiotics by Thin-

Layer Chromatography and Densitometry. Journal of Aoac International

2009;92(4):1068-1075.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 128: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

116

(163) Kaufmann A, Butcher P, Maden K. Determination of aminoglycoside residues

by liquid chromatography and tandem mass spectrometry in a variety of

matrices. Analytica Chimica Acta 2012;711:46-53.

(164) El-Attug MN, Hoogmartens J, Adams E, Van Schepdael A. Optimization of

capillary electrophoresis method with contactless conductivity detection for the

analysis of tobramycin and its related substances. Journal of Pharmaceutical

and Biomedical Analysis 2012;58:49-57.

(165) Adams E, Liu L, Dierick K et al. Neomycin: microbiological assay or liquid

chromatography? Journal of Pharmaceutical and Biomedical Analysis

1998;17(4-5):757-766.

(166) Oeltgen PR, Hamann SR, Blouin RA. Comparison of Radioimmunoassay (Ria)

and Enzyme-Immunoassay (Emit) for the Determination of Serum Gentamicin.

Clinical Chemistry 1980;26(7):970.

(167) Du M, Jones JR, Lanier J et al. Aminoglycoside suppression of a premature stop

mutation in a Cftr-/- mouse carrying a human CFTR-G542X transgene. J Mol

Med (Berl) 2002;80(9):595-604.

(168) Du M, Keeling KM, Fan L et al. Clinical doses of amikacin provide more

effective suppression of the human CFTR-G542X stop mutation than

gentamicin in a transgenic CF mouse model. J Mol Med (Berl) 2006;84(7):573-

582.

(169) Raz SR, Bremer MGEG, Haasnoot W, Norde W. Label-Free and Multiplex

Detection of Antibiotic Residues in Milk Using Imaging Surface Plasmon

Resonance-Based Immunosensor. Analytical Chemistry 2009;81(18):7743-

7749.

(170) Zhu Y, Son JI, Shim YB. Amplification strategy based on gold nanoparticle-

decorated carbon nanotubes for neomycin immunosensors. Biosensors &

Bioelectronics 2010;26(3):1002-1008.

(171) de Oliveira RC, Paschoal JAR, Sismotto M, Airoldi FPD, Reyes FGR.

Development and Validation of an LC-APCI-MS-MS Analytical Method for the

Determination of Streptomycin and Dihydrostreptomycin Residues in Milk.

Journal of Chromatographic Science 2009;47(9):756-761.

(172) Martos PA, Jayasundara F, Dolbeer J et al. Multiclass, Multiresidue Drug

Analysis, Including Aminoglycosides, in Animal Tissue Using Liquid

Chromatography Coupled to Tandem Mass Spectrometry. Journal of

Agricultural and Food Chemistry 2010;58(10):5932-5944.

(173) Bethune C, Bui T, Liu ML, Kay MA, Ho RJY. Development of a high-

performance liquid chromatographic assay for G418 sulfate (Geneticin).

Antimicrobial Agents and Chemotherapy 1997;41(3):661-664.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 129: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

117

(174) Stead DA. Current methodologies for the analysis of aminoglycosides. Journal

of Chromatography B 2000;747(1-2):69-93.

(175) Wang S, Xu B, Zhang Y, He JX. Development of enzyme-linked

immunosorbent assay (ELISA) for the detection of neomycin residues in pig

muscle, chicken muscle, egg, fish, milk and kidney. Meat Science

2009;82(1):53-58.

(176) Baasov T, Atia-Glikin D, Kandasamy J, Belakhov V; Aminoglycosides and uses

thereof in targeting genetic disorders. Israel patent WO 2012/066546 A1. 2011.

(177) Turnidge J. Pharmacodynamics and dosing of aminoglycosides. Infect Dis Clin

North Am 2003;17(3):503-28, v.

(178) Mehta M. PDR provides latest Food and Drug Administration-approved dosage

guidelines. Arch Intern Med 2001;161(21):2622.

(179) Sermet-Gaudelus I, Renouil M, Fajac A et al. In vitro prediction of stop-codon

suppression by intravenous gentamicin in patients with cystic fibrosis: a pilot

study. BMC Med 2007;5:5.

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 130: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

תקציר מחקר

י תכונות אנטיבקטריאליות פוטנטיות אמינוגליקוזידים מהווים משפחה של חומרי טבע בעל

אתר הפעולה . המשמשים בקליניקה כאנטיביוטיקות רחבות טווח לטיפול במגוון רחב של מחלות זיהומיות

הקטנה של המצוי בתת היחידה ( A-site-ה)של אמינוגליקוזידים בתא החיידקי הינו האתר המקודד

המובילות אל הריבוזום את ( tRNA)א "באתר זה מתרחשת הסלקציה למולקולות הטרנספר רנ. הריבוזום

כתוצאה מקישור אמינוגליקוזידים לריבוזום . חומצות האמינו המשמשות כאבני בניין לחלבון המתהווה

דבר שמוביל , נפגע מנגנון הסלקציה הריבוזומאלי ונכנסות שגיאות ראנדומליות בתהליך התרגום

חלבונים פגומים אלה גורמים בסופו . להצטברות של חלבונים שגויים ולא פונקציונאליים בתא החיידקי

מנגנון הפעולה של אמינוגליקוזידים בחיידקים נלמד כבר שנים רבות . של למותו של התא החיידקי

וזידים בריבוזום החיידקי כי אתר הקישור של אמינוגליק, למעשה ידוע. ובספרות קיים ידע רב אודותיו

מהווה מעין מתג מולקולארי המחליט על המשך או הפסקת תהליך התרגום לפי התאמת מולקולת

מתג זה מורכב משני שיירי . המקודד ליצירת החלבון( mRNA)א השליח "א לתבנית הרנ"הטרנספר רנ

. 1493-ו 1492אדנין שמורים מאוד אבולוציות הממוספרים

שני העשורים האחרונים הראו כי לאמינוגליקוזידים יש גם מספר אתרי מחקרים שבוצעו ב

מחקרים אלה הראו כי למעשה . בינהם גם ריבוזומים שמקורם ביצורים אאוקריוטים, קישור נוספים בטבע

לחלק מהנגזרות המשוייכות למשפחה יש גם יכולות תרפיוטיות נוספות כגון טיפול במחלות גנטיות

ק וטיפול במחלות זיהומיות שנגרמות כתוצאה מטפילים אאוקריוטים דוגמאת שמקורן במטוציית פס

למרות התוצאות הניסיוניות המבטיחות שהתקבלו (. אמבה טפילית)וגיארדיה ( שושנת יריחו)לישמניה

כמו גם המספר ההולך וגדל של מחקרים , בעבור שימוש באמינוגליקוזידים למטרות תרפיוטיות אלה

מנגנוני הפעולה של , ימוש באמינוגליקוזידים לטיפול במחלות אאוקריוטיות שונותקליניים הבוחנים ש

השימוש באמינוגליקוזים בקליניקה הוא יחסית , בנוסף. אמינוגליקוזידים באאוקריוטים עדיין אינם ידועים

. מוגבל בעיקר בעקבות תופעות הלוואי הקשות ורעילותם למשתמשים

רות סינטטיות של אמינוגליקוזידים לטיפול במחלות גנטיות בשנים האחרונות הוצעו מספר נגז

נגזרות אלה הן בעלות פעילות משופרת ורעילות מופחתת ביחס . י טפילים"ובמחלות זיהומיות הנגרמות ע

נגזרות אלה הינן גם סלקטיביות מאוד כלפי הריבוזום האאוקריוטי ובניגוד . לנגזרות הטבעיות הקיימות

מרבית הנגזרות הללו סונתזו במעבדת המחקר . כולות לשמש גם כאנטיביוטיקותלנגזרות הטבעיות לא י

לאור , עם זאת. שלנו והתוצאות הפרלימינריות שיש ברשותינו בעבור פעילותן ורעילותן הן מבטיחות

העובדה כי הידע הקיים ברשותינו בנוגע למנגנוני הפעילות של חומרים אלה במערכות אאוקריוטיות הוא

ד נוצר צורך לבצע מחקר שיתרום להבנה מעמיקה של מנגנוני הפעולה והאינטראקציות של מוגבל מאו

.אמינוגליקוזידים אלה עם אתרי המטרה האאוקריוטיים שלהם ברמה המולקולארית

י "במחקר שבוצע פענחנו את מנגנונים הפעולה של אמינוגליקוזידם במערכות אאוקריוטיות ע

הטכניקה . פיתרון המבנים התלת מיימדיים של אמינוגליקוזידים שונים קשורים לאתר המטרה שלהם

I © T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 131: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

כאשר במהלך המחקר גובשו , העיקרית בה השתמשנו לצורך המחקר היתה קריסטלוגרפיה של קרני איקס

עם ( חלקם אגוניסטים לאתר המטרה וחלקם אנטגוניסטים)קסים של אמינוגליקוזידים שונים קומפל

כאשר אתר המטרה הנחקר היה האתר . החלקים הרלוונטיים מריבוזומים של טפילים וריבוזומים אנושיים

במקביל . אנגסטרם 4.1-0.3המבנים הגבישיים נפתרו ברזולוציות אטומיות של . המקודד האאוקריוטי

ניסויים המבנים בוצעו גם מספר ניסויים ביוכימיים בהם נמדדו הפעילויות של אמינוגליקוזידים שונים ל

שילוב זה של מחקר מבני זה עם מחקר ביוכימי תרם לרכישת . ותאים אנושיים( לישמניה)על תאי טפילים

קישור שונה לאתר ידע רב בתחום והראה כי לאמינוגליקוזידים בעלי פעילות ביולוגית שונה יש תבנית

. הם משרים קונפורמציה שונה בעת קישורם, המטרה שלהם וכמו גם

הקרוי , נפתר בפעם הראשונה מבנה של קומפלקס של אמינוגליקוזיד, במסגרת עבודת המחקר

מבנה זה נפתר ברזולוציה של . עם אתר הפעולה שלו בטפיל אאוקריוטי הקרוי לישמניה( G418)נטיצין 'ג

ולמעשה הראה כי מנגנון הפעולה של אמינוגליקוזידים בלישמניה דומה למנגנון הפעולה אנגסטרם 6.2

מושרה בו , במבנה ניכר כי בעת קישור מולקולת האמינוגליקוזיד לאתר המטרה. שלהם בחיידקיים

קונפורמציה זו . קונפורמציה בה שני שיירי האדנין השמורים פונים אל מחוץ לסליל אליו הם משוייכים

א בעלת התאמה מלאה של כל שלושת "מושרית בריבוזום בעת קישור מולקולת טרנספר רנ כ"בד

קודון שעל גביה לשלושת הנוקליאוטידים המשוייכים לרצף הקודון המצוי -הנוקליאוטידים ברצף האנטי

קונפורמציה דומה מתקבלת גם בעת קישור אמינוגליקוזידים לריבוזומים . א השליח"ג מולקולת הרנ"ע

מחקרים מבניים . זאת למרות השינויים הרצפיים שבין אתרי הקישור בחיידק ובלישמניה, קייםחייד

וביוכימיים שבוצעו בחיידקים הראו כי קונפורמציה זו היא שתורמת להכנסת השגיאות הרנדומאליות

א באתר הקישור גם אם רצף האנטי"בתהליך התרגום בחיידקים תוך שהיא מייצבת מולקולות טרנספר רנ

. קודון שלהן אינו תואם לרצף הקודון

( HEK293)התוצאות המבניות שהתקבלו לוו גם בבדיקות עמידות של תרביות תאים אנושיים

וטפילי לישמניה לאמינוגליקוזידים שונים ובמסגרתם נמצא כי לאמינוגליקוזידים בעלי שייר של

יותר לשמש כחומרים תרפיוטיים יש יכולת טובה , הממוקמת בטבעת הראשונה, 2'הידרוקסיל בעמדה

י המבנים שנפתרו שהראו בפעם "תוצאות אלה נתמכו ע. לטיפול במחלות גנטיות ובמחלות טפיליות

הראשונה מדוע נגזרות אלה הינן בעלות פעילות משופרת ביחס לנגזרות המותמרות באופן שונה בעמדה

בעלות פעילות אנטילישמנייטית טובה הן הניסויים הביוכימיים שבוצעו הראו כי נגזרות שהן, בנוסף. זו

הדימיון הרב שבין אתר הקישור . י מוטציית פסק"גם יעילות יותר לטיפול במחלות גנטיות הנגרמות ע

מצביעות על כך כי מנגנון G418האנושי לזה של הטפילים כמו גם התוצאות המבניות שהתקבלו בעבור

יות המבוססות על קודוני פסק גם הוא דומה למנגנון הפעולה של אמינוגליקוזידים בתיקון מחלות גנט

. הכנסת השגיאות בחיידקים ובטפילים אאוקריוטיים

תוצאה מעניינת נוספת שהתקבלה במחקר היא שאמינוגליקוזידים שאינם משרים פעילות תיקון

, אאוקריוטים גנטי באדם או גורמים להכנסת שגיאות ראנדומאליות בתהליך התרגום בחיידקים וביצורים

II

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 132: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

אך ידועים כמעכבים חזקים של תהליך התרגום בכל מערכות החיים השרו קונפורמציה שונה לחלוטין

תוצאה זו למעשה מהווה הוכחה עקיפה למנגנוני הפעילות של . באתר הקישור בעת קישורם

ישור היא מכיוון שהיא מעידה שהקונפורמציה המושרית באתר הק, אמינוגליקוזידים בכל מערכות החיים

. זו שרלוונטית לצורך הפעילות הביולוגית ולאו דווקא עצם הקישור או חוזקו

, בנוסף להבנת המנגנונים המולקולאריים של פעילות אמינוגליקוזידים במערכות אאוקריוטיות

התוצאות הניסיוניות הביאו להבנה מעמיקה של תבניות הקישור של אמינוגליקוזידים שונים באתר

החשיבות של קבוצות פונקציונאליות בעמדות שונות לקיום הפעילות ולצורך הסלקטיביות הקישור ושל

אספקטים אלה של המחקר חשובים מאוד לפיתוח מושכל של נגזרות . של אמינוגליקוזידים למערכות הללו

.עתידיות של אמינוגליקוזידים בעלי פעילות משופרת ביחס לנגזרות האם

פותחה שיטה אימונית , יוכימיים שבוצעו במסגרת מחקר זהבנוסף למחקרים המבניים והב

י יצירה של נוגדנים "השיטה פותחה ע. לקביעה וכימות של אמינוגליקוזידים בדוגמאות ביולוגיות

פוליקלונאליים שהיו מסוגלים לזהות באפיניות גבוהה אמינוגליקוזידים סינטטיים שפותחו לצורך טיפול

שהופיע בכלל הנגזרות , י הזרקת הפטן גנרי"הנוגדנים יוצרו ע. ותבמחלות גנטיות ובמחלות טפילי

הנוגדנים שבודדו מהסרום של הארנבות שומשו ליצירת מבחן . לארנבות, הסנטטיות וצומד לחלבון נשא

היעילות והחזרתיות של השיטה הודגמה במספר מטריצות ביולוגיות . באריות 62דיאגנוסטי בצלחת של

שיטות . יושם בהצלחה על דגימות דם שנלקחו מעכברים שטופלו באמינוגליקוזידיםשונות וכמו גם המבחן

מכיוון שהן , אנליטיות מסוג הינן בעלות חשיבות רבה מאוד בפיתוח עתידי נוסף של הנגזרות התרפיוטיות

, בנוסף. מאפשרות ניטור יעיל ומהיר של הנגזרות בניסויים המבוצעים לבדיקת הנגזרות על בעלי חיים

לים שפותחו ניתן להשתמש גם להערכת אספקטים נוספים של פעילות הנגזרות כדוגמאת הערכת בכ

אספקטים אלה גם הם חשובים לפיתוח . חדירותן לתאים אנימאליים ומעקב אחרי פיזורם בתאים וברקמות

.ושיפור נגזרות עתידיות

. ערכות אאוקריוטיותבמחקר שבוצעו נחקרו מנגנונים הפעולה של אמינוגליקוזידים במ, לסיכום

תוצאות המחקר תרמו להבנת המרכיבים המולקולאריים הקשורים בסלקטיביות של אמינוגליקוזידים

מסויימים למערכות אלה ופתחו אפיקים חדשים ביכולת שלנו לפתח באופן מושכל נגזרות חדשות

יטה לניטור כמו גם פיתוח הש, הניסויים הביוכמיים שבוצעו במקביל למחקר המבני. ומשופרות

אמינוגליקוזידים ממטריצות ביולוגיות שונות תרמו רבות להבנה שלנו לגבי מנגנונים הפעולה של

תובנות אלה יוכלו לשמש בעתיד למחקר מקיף יותר ולפיתוח יעיל יותר . אמינוגליקוזידים באאוקריוטים

.של נגזרות חדשות למטרות תרפיוטיות אלה

III

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 133: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

נעם אדיר בפקולטה לכימיה' טימור באזוב ופרופ' פרופהמחקר בוצע בהנחיית

ברצוני להודות לטכניון ולמר סימור שוליך על התמיכה הכספית הנדיבה בהשתלמותי

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 134: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

הפעולה של אמינוגליקוזידים חקר מנגנוני

ככלי לפיתוח תרופות חדשות באאוקריוטים

חיבור על עבודת גמר

התואר לקבלת הדרישות של חלקי מילוי לשם

לפילוסופיה דוקטור

עמי-מורן שלו בן

לישראל טכנולוגי מכון - הטכניון לסנט הוגש

6340ג חיפה יוני "תמוז תשע

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary

Page 135: Technion – Israel Institute of Technologyלארשיל יגולונכט ןוכמ – ןוינכטה Technion – Israel Institute of Technology ןוינכטה תוירפס The Technion

הפעולה של אמינוגליקוזידים חקר מנגנוני

ככלי לפיתוח תרופות חדשות באאוקריוטים

עמי-מורן שלו בן

© T

echn

ion

- Isr

ael I

nstit

ute

of T

echn

olog

y, E

lyac

har C

entra

l Lib

rary