197
Synthesis and Applications of Polyfunctional Hydrogenated Pyridines and other Thiolated Ligands Stabilized Metal based Nanoparticles Submitted by Ayaz Anwar Dissertation for the Partial Fulfilment of the Degree of Doctor of Philosophy H. E. J. Research Institute of Chemistry International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan 2016

Dissertation for the Partial Fulfilment of the Degree of ...prr.hec.gov.pk/jspui/bitstream/123456789/3096/1/Ayaz_Anwar_Chemistry_2016_HSR_UoK...CV v CURRICULUM VITAE I was born in

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Synthesis and Applications of Polyfunctional Hydrogenated

Pyridines and other Thiolated Ligands Stabilized Metal

based Nanoparticles

Submitted by

Ayaz Anwar

Dissertation for the Partial Fulfilment of the Degree of

Doctor of Philosophy

H. E. J. Research Institute of Chemistry

International Center for Chemical and Biological Sciences,

University of Karachi, Karachi-75270,

Pakistan 2016

i

Certificate

To whom it may concern

It is certified that the thesis entitled, Synthesis and Applications of Polyfunctional

Hydrogenated Pyridines and other Thiolated Ligands Stabilized Metal Based

Nanoparticles, submitted to the Board of Advance Studies and Research, University of

Karachi, by Mr. Ayaz Anwar, satisfies the requirements for the Ph.D. degree in

Chemistry.

Dr. Muhammad Raza Shah,T.I. Research Supervisor

H. E. J. Research Institute of Chemistry,

International Center for Chemical and Biological Sciences,

University of Karachi, Karachi-75270, Pakistan.

ii

iii

DEDICATED TO

MY PARENTS AND MY FAMILY WHO ABETTED

ME IN THE BAFFLING COURSE OF MY

RESEARCH CORDIALLY AND EARNESTLY.

Acknowledgement

iv

ACKNOWLEDGEMENTS

I am thankful to Almighty Allah who grants me the power and courage tofulfil all my tasks. I

would like to dedicate and acknowledge this dissertation to my parents, and my siblings for

their constant prayers, encouragement and everlasting patience.

I am very thankful to Prof. Dr. Saleemuz Zaman Siddiqui (Late) founder HEJRIC, Patron in

Chief ICCBS, Prof. Dr. Atta ur Rahman FRS, (N.I, H.I., S.I., T.I.) and Director ICCBS, Prof. Dr. M.

Iqbal Choudhary(H.I., S.I., T.I.), for their wonderful leadership.

It is indeed my greatest pleasure to further extend my gratitude to my mentor and supervisor,

Dr. Muhammad Raza Shah (T.I.), for his constant support, encouragement and advices

throughout my Ph.D. studies as well as in my personal issues. He is a source of inspiration for

me to accomplish this task and no doubt my role model. His valuable efforts in encouraging me

to achieve my goals with a helping and kind heart made it possible for me. I cannot thank him

enough for his support throughout every day of my stay in his wonderful research group.

I would like to extend my thanks to all the collaborating groups. Dr. Massimo F. Bertino of

Virginia Commonwealth University USA, Dr. Yousuf Aqeel, and Dr. Naveed Ahmed Khan of

Aga Khan University, Dr. Fatima Basha, and extremely thankful to Dr. Abdul Hameed of

ICCBS for his guidance and mentorship in the synthesis of dihydropyridine derivatives for their

valuable support in my research. Very special thanks to all the faculty members of ICCBS who

laid my research foundation during the course work. I would also like to acknowledge all the

technical and non-technical staff of ICCBS.I am very much thankful to Mr. Amir for his

support in timely arrangements of research materials.

I would like to acknowledge and pay my special thanks to all my lab fellows and friends in

particular, Dr. Nadeem, Dr. Qamar, Dr. Akhtar, Dr. Zahid, Dr. Farrukh, Kiramat, Burhan, Dr.

Ateeq, Mehdi, Mumtaz, Tahir, Ambreen, Anwar Shamim, Farid, Ishfaq, Afifa, Wasia, Noor,

Azeem, Saira, Faiza, Dania, Sadia, Sadia Khan, Madiha, Imkaan, Imdad.

Special thanks to my friends Hamid, Umair, Afifa, Farid, Ishfaq, Zahid, M. Salman, Mateen,

Junaid, Saquib, Comail, Ateeq, Shabbir, Nusrat, all class fellows, all LPO members, and my

cousins Waqar, Danish, Uzair,and many more for their moral supports and appreciations.

At last but not the least, I would like to thanks all the members of Mass, NMR, and Analytical

instrumental lab of ICCBS.

Ayaz Anwar, Karachi, 2015

CV

v

CURRICULUM VITAE

I was born in a small extended town of Karachi, Pakistan called Landhi on the 4th of

January 1988. I gained my school education from a nearby school named Sohail

Academy. After Matric, I joined Urdu Science College Gulshan e Iqbal for F. Sc. and

somehow managed to pass the intermediate education. I then took my studies a little

more serious, and I realized that Chemistry is the subject which fascinates me and I

decided to learn the Chemistry of the highest levels right then. So I graduated in

Chemistry, Mathematics and Physics from Jamia Millia Government Degree College

Karachi in 2007. Later on, I joined the University of Karachi for postgraduate studies in

Organic Chemistry, where received M.Sc. degree with distinction on 2010. After the

exams, I had a great opportunity to join the summer internship program of ICCBS, where

I had my first exposure of Chemistry research. In March 2011, I joined the H.E.J.

Research Institute of Chemistry, International Centre for Chemical and Biological

Sciences, University of Karachi, for Ph.D. studies in Chemistry. I enjoyed research

activities in the field of Organic Synthesis and nanotechnology under the supervision of

Dr. Muhammad Raza Shah, mainly devoted to the nanoparticles based studies of metal-

coated synthetic ligands for enhanced photophysical and biological applications. I feel

myself very lucky to reach this day. It has been an extraordinary life so far, I enjoy

playing and watching football, watching movies and listening music.

Table of Contents

Certificate ...................................................................................................................... i Dedication ................................................................................................................... iii Acknowledgement ...................................................................................................... iv Personal Introduction ................................................................................................... v List of figure ................................................................................................................ x List of schemes ........................................................................................................... xv List of tables ............................................................................................................... xvi List of abbreviations ................................................................................................. xvii Summary ................................................................................................................. xviii Khulasa ..................................................................................................................... xix

PART I ........................................................................................................................ 1

1.1.1 Dihydropyridines ............................................................................................... 1

1.1.2 Porphyrin............................................................................................................ 5

1.1.2.1 Nomenclature .................................................................................................. 6

1.1.2.2 Synthetic Methodologies of Porphyrins.......................................................... 7

1.1.3 Thiolated Cationic Heterocyclic Ligands for Stabilization of Metal Nanoparticles ............................................................................................................ 12

1.2 RESULTS AND DISCUSSION ........................................................................... 13

1.2.1 Synthesis and Applications of Polyfunctional Hydrogenated Pyridines from Acetophenone Derivatives ........................................................................................ 13

1.2.1.1 Urease Activity and Structural Activity Relationship .................................. 15

1.2.1.2 Anticancer Activity and Structural Activity Relationship ............................ 17

1.2.1.3 DPPH Radical Scavenging Activity ............................................................. 17

1.2.2 Synthesis of Mannose Conjugated Porphyrin .................................................. 20

1.2.2.1 Synthesis of Tetraphenylporphyrin (TPP) (7)............................................... 20

1.2.2.2 Nitration of TPP (27) .................................................................................... 21

1.2.2.3 Mechanism of Nitration of Meso-H2TPP ..................................................... 22

1.2.2.4 Reduction of nitro group of MNPP ............................................................... 23

1.2.2.5 Mechanism of Nitro group of Reduction ...................................................... 24

1.2.2.6 Conjugation of MAPP with Mannose ........................................................... 25

vi

1.2.2.7 Antibacterial Study of Mannose Conjugated Porphyrin via Photochemotherapy................................................................................................... 26

1.3 EXPERIMENTAL ................................................................................................ 33

1.3.1 General Procedure for the Synthesis of Polyfunctional DHP Derivatives ...... 33

1.3.2 Urease Inhibition Assay (In vitro) ................................................................... 38

1.3.3 Anticancer Activity Assay (In vitro) ................................................................ 38

1.3.4 DPPH (1,1-Diphenyl-2-Picryl Hydrazyl) Free Radical Scavenging Assay (In vitro) .......................................................................................................................... 39

1.3.5 Acanthamoeba castellanii and Microvascular Endothelial Cells Cultures ...... 40

1.3.6 Synthesis of Mannose Conjugated Porphyrin .................................................. 40

1.3.7Amoebicidal Assays.......................................................................................... 41

1.3.8 Cytotoxicity assays of Host Cells .................................................................... 42

1.3.9 Excystation Assays .......................................................................................... 42

1.3.10 Synthesis of thiolated heterocyclic ligands .................................................... 43

References ................................................................................................................... 47 PART II ..................................................................................................................... 55

SYNTHESIS OF METAL NANOPARTICLES AND THEIR APPLICATIONS .... 55 2.1 INTRODUCTION ................................................................................................ 55

2.1.1 Introduction of Nanotechnology ...................................................................... 55

2.1.2 Fullerenes ......................................................................................................... 55

2.1.3 Carbon Nanotubes ............................................................................................ 56

2.1.4 Nanoparticles ................................................................................................... 57

2.1.5 Nanoclusters ..................................................................................................... 59

2.1.6 Synthesis of Nanoparticles and Nanoclusters .................................................. 60

2.1.6.1 Bottom-up Approach .................................................................................... 60

2.1.6.2 Top-down Approach ................................................................................... 60

2.1.7 Dispersity in Nanoscale Synthesis ................................................................... 60

2.1.8 Applications of Nanoparticles.......................................................................... 60

2.1.9 Nanoclusters in Biomedical Sciences .............................................................. 61

2.1.9.1 Biological Imaging........................................................................................ 61

2.1.9.2 Biodetection .................................................................................................. 62

vii

2.1.10 Chemosensing of Cu(I) .................................................................................. 63

2.1.11 Chemosensing of Fe(III) ................................................................................ 63

2.1.12 Chemosensing of Pd(II) ................................................................................. 64

2.1.13 Chemosensing of Antibiotics ......................................................................... 66

2.2 RESULTS AND DISCUSSION ........................................................................... 69

2.2.1.1 Synthesis and Characterization of Bisphosphine Gold Nanoclusters ........... 69

2.2.2 Atomic Force Microscopic guided detection of Copper in real samples and T-lymphocytes with Pyridinium Thioacetate capped Silver Nanoparticles ................. 82

2.2.2.1 Synthesis of 4-formylpyridiniumpropylthioacetate Coated Silver Nanoparticles (4-Py-AgNPs) .................................................................................... 82

2.2.2.2 Characterization of 4-Py-AgNPs .................................................................. 82

2.2.2.3 Chemosensing of Cu(I) by 4-Py-AgNPs....................................................... 85

2.2.2.4 Intracellular Cu(I) detection in T-lymphocytes: ........................................... 96

2.2.2.5 Chemosensing of 6-AminopenicillanicAcid by 4-Py-AgNPs .................... 104

2.2.2.6 Antimicrobial Activity of 4-Py-AgNPs ...................................................... 112

2.2.3 Synthesis of Pyrazinium Thioacetate Gold Nanoparticles Pyr-AuNPs and their Applications ............................................................................................................ 117

2.2.3.1 Synthesis of Pyr-AuNPs ............................................................................. 117

2.2.3.2 Characterization of Pyr-AuNPs .................................................................. 117

2.2.3.3 Photophysical Applications of Pyr-AuNPs ................................................. 118

2.2.4 A New Pefloxacin Chemosensor Based on Thioacetate Functionalized DMAP coated Gold Nanoparticles ...................................................................................... 135

2.2.4.1 Synthesis of Dimethylaminopyridinium Propylthioacetate (D2) Stabilized Gold Nanoparticles (D2-AuNPs) ............................................................................ 135

2.2.4.2 Stability of D2-AuNPs ................................................................................ 137

2.2.4.3 Exploration of Chemosensing Potential of D2-AuNPs .............................. 139

2.2.4.4 Effect of the pH on the Stability of ................... D2-AuNPs and Pef Response................................................................................................................................. 140

2.2.4.5 FT-IR Spectral Analysis ............................................................................ 141

2.2.4.6 AFM investigation of the interaction between D2-AuNPs and Pef ......... 141

2.2.4.7 Analytical Performance of the Pef Nanosensor .......................................... 142

viii

2.2.4.8 Interference Study ....................................................................................... 144

2.2.4.9 Analysis of Pef in Real Samples ................................................................. 145

2.3 EXPERIMENTAL .............................................................................................. 147

2.3.1 Materials ........................................................................................................ 147

2.3.2 Instruments ..................................................................................................... 147

2.3.3 Synthesis of Bisphosphines Coated Gold Nanoclusters ................................ 148

2.3.4 Synthesis of 1-(3-(acetylthio)propyl)-4-formylpyridin-1-ium (4-Py) Coated Silver Nanoparticles (4-Py-AgNPs) ........................................................................ 148

2.3.5 Synthesis of 1-(3-(acetylthio)propyl)pyrazin-1-ium (Pyr) Coated Gold Nanoparticles (Pyr-AuNPs) .................................................................................... 148

2.3.6 Synthesis of 1-(3-(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium (D2) Coated Gold Nanoparticles (D2-AuNPs) ................................................................ 149

2.3.7 Synthesis of Chlorhexidine (CHX) Coated Gold Nanoparticles (CHX-AuNPs)................................................................................................................................. 149

2.3.8 Synthesis of Neomycin (Neo) Coated Silver Nanoparticles (Neo-AgNPs)... 149

2.3.9 Synthesis of Guanidinobutyric Acid (GBA) Coated Silver Nanoparticles (Neo-AgNPs) .................................................................................................................... 149

2.3.10 Cell Incubation ............................................................................................. 150

2.3.11 Cell Treatment ............................................................................................. 150

2.3.12 MTT Based Cytotoxicity Assay................................................................... 150

2.3.13 Minimum Inhibitory Concentration by Agar Well Diffusion Method ........ 151

2.3.14 Atomic Force Microscopy Imaging ............................................................. 151

REFERENCES ......................................................................................................... 153 Conclusion ............................................................................................................... 177 Publications .............................................................................................................. 178

ix

x

List of Figures

Figure: 1.1 Structures of Nifedipine (1) and Lercanidipine (2) 1

Figure: 1.2 Comparative structures of our DHPs and some reported analogues

with drugs

4

Figure: 1.3 The tautomerization of porphyrins 6

Figure: 1.4 Typical structure of simplest porphyrin- porphyne 6

Figure: 1.5 Fischer (left) and IUPAC (right) nomenclature systems for

porphyrins

7

Figure: 1.6 X-rays crystallographic structures of compounds 9 and 11 14

Figure: 1.7 NMR spectrum of MNPP (27) 22

Figure: 1.8 EI-MS spectrum of MAPP (28) 24

Figure: 1.9 NMR spectrum of mannose conjugated porphyrin (29) 25

Figure: 1.10 Mannose-conjugated porphyrin (mann. por.) exhibited

amoebicidal effects against A. castellanii.

27

Figure: 1.11 A. castellanii was incubated with and without 50 µM mannose

conjugatedporphyrin, and representative images were obtained at 100 x (A)

and at400 x (B) magnification using a phase-contrast inverted microscope.

28

Figure: 1.12 (A). A. castellanii (5 x105) preincubated with and without 50

µM mannose-conjugated porphyrin (mann. por.) and exposed to light. (B). A.

castellanii (5 x105) were coincubated with human BMEC and 50µM

mannoseconjugatedporphyrin and exposed to light.

30

Figure: 1.13 Mannose-conjugated porphyrin inhibited excystation of A.

castellanii cysts. A. castellanii cysts were scraped from nonnutrient agar

plates, incubated with 50µM mannose porphyrin, and exposed to light.

31

Figure: 2.1 Structure of a Fullerene 56

xi

Figure: 2.2 Depiction of CNTs 57

Figure: 2.3 (A) 1,6-bis(diphenylphosphino)hexane denoted by L6, (B)

Xantphos denoted by LAr, (C) Binap. LBn, (D) Biphenyl denoted by LBp,

(e) Diphenylphosphinoacetylene denoted by LAc

69

Figure 2.4 UV-Visible Spectrum of L6 Au Clusters in all solvents 71

Figure: 2.5 MALDI spectra of L6 obtained in all solvents 72

Figure: 2.6 UV-Vis spectrum of LAr Au Clusters in all solvents used 73

Figure 2.7 MALDI Spectral Ananlysis of LAr Ligated Au Clusters in

different solvents

75

Figure: 2.8 UV-Vis Spectra of LAc Au Clusters in all Solvents 76

Figure: 2.9 MALDI Spectral Ananlysis of LAc Ligated Au Clusters in

different solvents

77

Figure: 2.10 UV-Vis Spectra of LBn Au Clusters in all solvents 78

Figure: 2.11 MALDI Analysis of LBn Au Clusters in all Solvents 79

Figure: 2.13 MALDI Analysis of LBp Au Clusters in all Solvents 81

Figure: 2.14 UV-visible spectra of the AgNPs 83

Figure: 2.15 UV-visible spectra of the AgNPs solution at room temperature

and boiling point

84

Figure: 2.16 UV-visible spectra of salt addition stability of AgNPs 85

Figure: 2.17 A) UV-visible spectra of metal cations after titrations with

nanoparticles. B) Bar graph representation of metal cations after titrations

with nanoparticles at λmax

86

Figure: 2.18: A) FT-IR spectrum of 4-Py AgNPs 87

Figure: 2.18 B) FT-IR spectrum of 4-Py AgNPs after treating with Cu(I) 88

Figure: 2.19 a) Topographical AFM image of 4-Py AgNPs b) Topographical

AFM image of AgNPs after mixing with Cu(I)

89

xii

Figure 2.20 Kinetic studies after interaction of Cu(I) with4-Py AgNPs 90

Figure: 2.21: Effect of pH on the selective recognition of Cu(I) by AgNPs. 91

Figure: 2.22 A) Selective recognition of Cu(I) by 4-Py AgNPs at different

concentrations B) Linear regression curve obtained after treating varies

concentration of Cu(I) with 4-Py AgNPs.

92

Figure: 2.23 A) Absorption spectra of 4-Py AgNPs+Cu(I) in presence of

other competing metal cations B) bar graph representation Absorption spectra

of 4-Py AgNPs+Cu(I) in presence of other competing metal cations

93

Figure: 2.24 Complexation ratio between 4-Py AgNPs and Cu(I) 94

Figure 2.25 UV-visible spectra of 4-Py AgNPs recorded in presence Cu(I)

spiked in (A) Human plasma (B) Tap water

95

Figure: 2.26 A) T cell (Control) 98

Figure: 2.26 B) 0.5 μM 4-Py AgNPs treated T cell 98

Figure: 2.26 C) 0.5 μM 4-Py AgNPs treated T cell with 5 μMCu (I), cell

surface is shrinked due to interactions.

99

Figure: 2.26 D) 0.5 micromolar 4-Py AgNPs treated T cell with 10

micromolar Cu (I)

100

Figure: 2.27 A) 1 micromolar 4-Py AgNPs treated T cell with 5 micromolar

Cu (I)

101

Figure: 2.27 B) 1 micromolar 4-Py AgNPs treated T cell with 30 micromolar

Cu (I)

101

Figure: 2.28 A) 10 micromolar 4-Py AgNPs treated t cell 102

Figure: 2.28 B) 10 micromolar 4-Py AgNPs treated t cell with 5 micromolar

copper

102

Figure: 2.28 C) 10 micromolar 4-Py AgNPs treated t cell with 50 micromolar

copper

103

Figure: 2.29 Drugs screening study 106

xiii

Figure: 2.30 Morphological change in the nanoparticles after interaction with

6-APA

107

Figure: 2.31 Effect of pH on the stability of 4-Py AgNPs and accumulation of

nanoparticles with 6-APA

108

Figure: 2.32 A) UV-visible spectra of concentration study, B) Calibration

curve and C) Job’s plot

110

Figure: 2.33 Effect of interfering drugs on 6-APA quenching 111

Figure: 2.34 6-APA quenching in Human blood plasma sample 112

Figure: 2.35 MIC of ligand in comparison with ligand stabilized 4- Py-

AgNPs in the presence of Cu(I).

114

Figure: 2.36 A) E. coli Control. B) E. coli after treating Cu(I) 116

Figure: 2.37 Ligand treated E. coli at A) 4 hours. B) 8 hours 116

Figure: 2.38 4- Py-AgNPs treated E. coli at A) 4 hours. B) 8 hours 116

Figure: 2.39 UV-visible spectrum of Pyr-AuNPs 117

Figure: 2.40 UV-visible spectra of metal salts screening 119

Figure: 2.41 Buffer stability of AuNPs with and without Fe(III) 120

Figure: 2.42 Comparative FT-IR spectra of Pyr-AuNPs before and after

mixing of Fe(III)

121

Figure: 2.43 AFM images of AuNPs alone and of AuNPs treated with Fe(III) 122

Figure: 2.45 Effect of interfering metal salts on the Fe(II) sensing 124

Figure: 2.46 Fe(III) sensing in drinking water and human blood plasma 126

Figure: 2.47A) Pictorial display of colorimetric sensing of Pd(II), B) UV-

visible spectrum of salt screening

128

Figure: 2.48 pH stability of AuNPs with and without Pd(II) 129

Figure: 2.49 A) AFM image of AuNPs, B) AFM image of AuNPs after

mixing with Pd(II)

130

xiv

Figure: 2.50 A) UV-visible spectral profile of the concentration study, inset

represents calibration curve, B) Job’s plot

132

Figure: 2.51 Effect of interfering metal ions on Pd(III) sensing 133

Figure: 2.52 UV-visible spectra of Pyr-AuNPs after mixing with A) tap

water, B) Human blood plasma spiked with Pd(II)

134

Figure: 2.53 A) UV-Visible spectrum of the AuNPs stabilized with D2, B)

AFM image of the D2 capped AuNPs, C) Size distribution histogram of D2-

AuNPs

137

Figure: 2.54 A) Effect of heating on SPB, B) Aggregation of D2-AuNPs

induced by various concentration of NaCl

138

Figure: 2.55 UV-Visible spectral profile generated after mixing drugs with

D2-AuNPs

139

Figure: 2.56 Effect of pH on the interaction of D2-AuNPs with Pef 140

Figure: 2.57 AFM image of the D2-AuNPs -Pef 1:1 mixture 142

Figure: 2.58 A) Concentration study of Pef with D2-AuNPs, B) Calibration

curve at 700 nm C) Job’s plot for binding ratio

144

Figure: 2.59 Effect of competing drugs on Pef response towards nanosensor 145

Figure: 2.60 UV-visible spectra of Pef recognition in (A) tap water (B)

Human blood plasma

147

xv

LIST OF SCHEMES

Scheme 1.1 Synthesis of meso-H2TPP (7) by Alder-Longo Method. 9

Scheme 1.2 Synthesis of meso-H2TPP (7) by Lindsey Method. 10

Scheme 1.3 Synthesis of meso-H2TPP(8) by MacDonald method. 10

Scheme 1.4 Proposed Mechanism of synthesis of meso-H2TPP. 11

Scheme 1.5 Synthetic methodology of the target DHPs. 13

Scheme 1.6 Plausible mechanism for the dimerization of alkylidene

malononitrile to give DHP.

15

Scheme 1.7 Synthesis of meso-H2TPP (7). 20

Scheme 1.8 Synthesis of MNPP (27). 21

Scheme 1.9 Mechanism of nitration of meso-H2TPP. 22

Scheme 1.10 Reduction of MNPP into MAPP. 23

Scheme 1.11 Proposed mechanism of reduction. 24

Scheme 1.12 Synthesis of Mannose conjugated Porphyrin (29). 25

Scheme 1.13 Synthesis of 1-(3-(acetylthio) propyl)-4-formylpyridinium (4-

Py)(31)

43

Scheme 1.14 Synthesis of 1-(3-(acetylthio)propyl)pyrazin-1-ium (Pyr) (33) 44

Scheme 1.15 Synthesisof1-(3-(acetylthio)propyl)-4-(dimethylamino)

pyridin- 1-ium (D2) (34)

45

xvi

LIST OF TABLES

Table 1.1 In vitro urease activity of DHPs. 16

Table 1.2 Anticancer activity of DHPs against PC3 and HeLa Cell line. 18

Table 1.3 DPPH free radical scavenging activity. 19

Table 1.4 Structures and spectroscopic data of synthesized DHPs. 34

Table 2.1 Zone of inhibition data at multiple dose of Cu(I) accumulation. 113

xvii

LIST OF ABBREVIATIONS

AuNPs Gold Nanoparticles

AgNPs Silver Nanoparticles

SPB Surface Plasmon Band

AFM Atomic Force Microscopy

DHP Dihydropyridine

HMDS Hexamethyl disilazane

TBAF Tetra-n-butyl ammonium fluoride

4-Py 1-(3-(acetylthio)propyl)-4-formylpyridinium

Pyr 1-(3-(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium

D2 1-(3-(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium

MALDI Matrix Assisted Laser Desorption/Ionization

FTIR Fourier Transform Infrared

IR Infrared

ESI Electrospray Ionization

EI Electron Impact

MS Mass spectrometry

NMR Nuclear Magnetic Resonance

PE Petroleum Ether

EA Ethyl acetate

DMSO Dimethyl Sulfoxide

NaOH Sodium Hydroxide

nps Nanoparticles

APA 6-Aminopenicillanic acid

Pef Pefloxacine

xviii

SUMMARY

This Ph. D. dissertation, deals with the synthesis of heterocyclic thiolated ligands,

stabilization of metal nanoparticles and their photophysical and biological applications. The

ligands explored in this study include derivative of Pyridines, Dihydropyridines, Pyrazine,

and Porphyrin. Chemical synthesis are monitored and characterized by1HNMR, Mass

spectrometry, FT-IR spectrophotometry, while the nanomaterials were studied by UV-visible

spectrophotometry, and AFM. The antimicrobial potential of synthesized ligand and

nanoparticles were also studied comprehensively. As a summary of the research conducted

during the course of Ph. D., a series of novel dihydropyridine derivative was synthesized and

their cytotoxicity, enzyme inhibition and antioxidant activity were studied. Para-methoxy

dimer of alkylidene malononitrile is found to be most active in in vitrourease inhibition

assay, while para-bromo analogue is found to be significantly cytotoxic against two cell lines.

Moreover, 1-(3-(acetylthio) propyl)-4-formylpyridinium (4-Py), 1-(3-(acetylthio)propyl)

pyrazin-1-ium (Pyr), 1-(3-(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium (D2) were

synthesized as thiolated capping agents for stabilization of metal nanoparticles particularly

Silver and Gold. 4-Py coated AgNPs were successfully synthesized and used as chemosensor

for detection and quantification of Cu(I) ions and 6-Aminopenicillanic acid detection in

aqueous media in the presence of other interfering species. Pyr capped AuNPs were

synthesized, which colorimetrically sensed Pd (II)in the presence of other metal ions by

inducing hypochromic shift in the surface plasmon band, and Fe(III) ions by quenching of

SPR band. D2 stabilized gold nanoparticles were synthesized, which exhibited selective

recognition of Pefloxacin mesylate by showing a substantial red shift in the SPR band. The

antibacterial potential of 4Py-AgNPs was also explored and compared with uncoated ligand

against E. coli, and the morphological studies were carried out via AFM. Silver conjugated 4-

Py displayed two times enhanced anti-E. coli activity as compared to pure compound, while

the intracellular Cu(I) detection was also successfully achieved, which can be used to reduce

the copper poisoning in biological systems. In another study, a unique selective

Photochemotherapic agent mannose conjugated porphyrin was synthesized and used against

the Acanthamoeba via in vitro studies. Three antibiotics Chlorhexidine, Neomycin, and

guanidine butyric acid were also decorated on metal nanoparticles surface to study their

antibacterial activities.

xix

Part I

1

CHAPTER 1

SYNTHESIS OF HETEROCYCLIC ORGANIC LIGANDS AND

THEIR BIOLOGICAL APPLICATIONS

1.1 INTRODUCTION

1.1.1 Dihydropyridines

Alkylidenemalononitriles are important targets and intermediates compounds in synthetic

and bioorganic chemistry. These are synthesized from acetophenone and malononitrile

derivatives via Knoevenagel condensation (Barnes, Haight et al. 2006) and are often used

in the preparation of many medicinally vital heterocyclic compounds. Pyridines are also

an important class of the heterocyclic compounds, which are commonly used in many

fields including medicinal chemistry, agro chemicals etc. (Bogdanowicz-Szwed and

Krasodomska 2006). Pyridine ring is the core in many known natural products and

medicinally important compounds, for example, nifedipine and its different derivatives

(Johnson and Li 2007) (Fig.1.1). These compounds showed significant biological

activities against cardiovascular diseases and act as calcium antagonist (Lichitsky,

Dudinov et al. 2001). Many research groups are recently involved in the synthesis of

useful pyridine derivatives owning to their significant biological potential. Based on these

properties and recent research reports, we aimed to develop an efficient and cost-effective

method for the synthesis of potentially useful pyridine derivatives. It is predicted that

preparation of poly functional 5, 6-pyridine derivatives would carry significant biological

activities.

1 2

Figure: 1.1 Structures of Nifedipine (1) and Lercanidipine (2)

Part I

2

Readily available alkylidenemalononitriles were selected as suitable precursor due to the

reactive nature of the dicyanoolefin and the acidic nature of the methyl group for the

dimerization reaction to synthesize desired 5,6-dihydropyridine derivatives 9-26

(Table1.4). Previously, (Dunkel, Hess et al. 1997) reported the synthetic strategy to

produce different dicyano substituted dimers by using DIMCARB (dimethylamine

carbondioxide complex), while (Abdelrazek, Kassab et al. 2010) carried out the

dimerization of phenyl alkylidenemalononitrile in the presence of NaOEt in ethanol.

Alkylidenemalononitriles can be formed by knoevenagel condensation of acetophenones

and malononitrile. There are several reported methods for this purpose (Knoevenagel

1898; Barnes, Haight et al. 2006). For example:

However, we have developed a novel one-pot solvent-free methodology for the synthesis

of alkylidenemalononitrile and their subsequent conversion into DHP derivative via

dimerization. The reaction is carried out in neat TBAF.3H2O (Tetrabutylammonium

fluoride trihydrate) to prepare highly functionalized compound 9-26 (Table 1.4). These

conditions are green as no organic solvent is used, eco-friendly, facile and rapid. We have

explored this optimized method on a variety of different substituted alkylidenemalononitriles

analogues and synthesized 16 different dihydropyridine derivatives.

Part I

3

TBAF is commonly used for deprotection of silyl group from silyl ethers in organic

reactions (Corey and Snider 1972). However, recently several research groups have

explored many other applications of this reagent, for example, nucleophilic substitution

of fluoride to synthesize organo-fluoro compounds (Cox, Terpinski et al. 1984),

cyclization reactions for the synthesis of different heterocyclic rings systems (Hiroya,

Jouka et al. 2001; Yasuhara, Suzuki et al. 2002), oxidation reaction (Chung, Moon et al.

2006), click chemistry (Amantini, Beleggia et al. 2004) and [4+2]-cycloaddition (Alden-

Danforth, Scerba et al. 2008) etc. We for the first time explored TBAF.3H2O as a base for

the dimerization of alkylidenemalononitriles 9-26 (Table 1.4) under solvent-free

conditions. Since pyridine derivatives are an important class of N-heterocyclic

compounds which are reported to exhibit a wide range of biological activities, including

antifungal (Hargreaves, Pilkington et al. 2000), antibacterial (Malicorne, Bompart et al.

1991) and anticancer (Szökő and Tábi 2010). In this perspective, we evaluated a series of

DHPs as urease inhibitor, anticancer, and antioxidant agent.

The development of urease inhibitors is directly a step towards the medicinally important

antiulcer agents. Urease enzyme is clinically used for the diagnosis of pathogens in the

gastrointestinal and urinary tract. It has been associated with the several infectious

diseases, like stomach cancer, stones and peptic ulcer formation etc. (Burne and Chen

2000). Urease is also involved in other pathogenesis like urolithiasis, hepatic come, oral

cavity infections, and urinary catheter encrustation (Mobley, Island et al. 1995). Since

Urease metal part is consist of Nickle, our polyfunctional DHPs especially substituted

with electron donating groups are an ideal candidate to interact with the enzyme and

cause its inhibition.

On the other hand, Cancer, an unrestrained aggregation of abnormal cells is one of the

major causes of human mortality in the world. The available doxorubicin IV and related

compounds chemotherapies are encountered with some major concerns such as

cardiotoxicity (Menna, Gonzalez Paz et al. 2012) and multidrug-resistance (MDR). One

of the main causes of MDR mainly occur due to the over expression of multidrug

resistance protein efflux pump, which extrude the drugs from the cell (Volm, Mattern et

al. 1991; Talelli, Morita et al. 2011). Toyota et al., reported the synthesis of 1,4-

Part I

4

dihydropyridine, calcium channel antagonist, (Structure 3, Fig. 1.2) derivatives and used

them in combination with commercially doxorubicin IV. Because, multiple resistant cells

enhance the membrane transportation, in particular the excretion process which lower the

drug concentration in the cells. So, by increasing the anticancer drug concentration in

cancer cells through inhibiting extracellular excretion process of cell, their method

displayed quite interesting outcome (Toyota, Shinkai et al. 1993). Inspired by their work

and of (Abadi, Ibrahim et al. 2009) antitumor investigations in drug resistant cancer cells,

we screened our polyfunctional dihydropydridine compounds (Structure 5) for anticancer

activity (Fig. 1.2). And the results showed a wide range of both Urease inhibition and

cytotoxic activities depending upon the substituents present on the phenyl rings.

Figure: 1.2 Comparative structures of our DHPs and some reported analogues with drugs

Moreover, on the basis of very deep understanding of antioxidant research studies, it is

now a well-known fact that free radicals generated due to oxidative biochemical

processes cause many biological oxidative damages, including protein denaturing,

mutagenesis and several other degenerative pathological events (Beckman and Ames

Part I

5

1998; Cui, Kong et al. 2012). These damages are responsible for aging, diabetes, cancer

and asthma (Meyer, Heinonen et al. 1998; Hunt, Lester et al. 2001; Martínez-Martínez,

Razo-Hernández et al. 2012). A literature review shows that various examples of

antioxidant agents bearing heterocyclic aromatic rings are reported to inhibit cellular

oxidative damages (Rice-Evans, Miller et al. 1996; Villaño, Fernández-Pachón et al.

2007). The polyphenols are known to have a tremendous antioxidant potential, and

hundreds of species of polyphenols display antioxidant activity in vitro, but their

antioxidant role in in vivo studies are found to be significantly low and matter of concern

which give rise to the quest of more efficient antioxidant agents (Williams, Spencer et al.

2004; Ajitha, Mohanlal et al. 2012). Although all oxidative reactions are not less

important for living cell processes some of them are vital, but the main problem of these

are the formation of free radicals in such reactions, which due to their high reactivity

initiate many undesired chain reactions in cell, which can ultimately lead to cell damage

and/or death. The mode of action of an antioxidant compound can be generally regarded

as an electron contributor to free radical species to convert them into harmLess

molecules, so that the cells are prevented from oxidative damage (Holiman, Hertog et al.

1996; Moure, Cruz et al. 2001). So, the development of biologically benign radical

scavengers to inhibit unwanted oxidative reactions is an important field of interest for

antioxidant researchers.

1.1.2 Porphyrin

Porphyrins (Greekoriginated means “purple, scarlet”) are very significant class of natural

occurring molecules. Several biologically significant molecules for example, vitamin B-

12, heme, and chlorophylls contain porphyrin in their basic skeleton (Willstatter and Stoll

1928; Boucher and Katz 1967; Lemberg and Barrett 1973; Hodgkin 1979; Milgrom

1997). Porphyrin macrocyle contains 22 π-electrons, among which only 18 are found to

actually participate in any one delocalization pathway as shown in Fig. 1.3. It follows

Hückel’s [4n+2] rule for aromaticity, where n = 4. While the other four electrons are

situated outside the delocalization pathway are on the two double bonds opposite to each

other at pyrrole rings.

Part I

6

Figure: 1.3 The tautomerization of porphyrins

As a typical aromatic system, porphyrin is able to undergo electrophilic aromatic

substitution reactions (EAS), such as nitration, halogenation and formylation at any

unsubstituted meso- and/or β-pyrrolic positions (Smith and Falk 1975). However, the

quaternary α-pyrrolic carbons are almost inert, and do not give the substitution products.

While the two double bonds outsidethe delocalization pathway are susceptible to

reductions and oxidations, for example: catalytic hydrogenation, reduction with diimide,

and hydroboration.

Figure: 1.4 Typical structure of simplest porphyrin- porphyne

1.1.2.1 Nomenclature

According to the older nomenclature (Fischer system), the meso positions are labeled by

a Greek letters system, and the four pyrrolic sub-unit rings are also labeled with the

capital letters A, B, C, and D. However the more modern IUPAC system identifies every

carbon in the macrocyclic ring and it also numbered the carbon of the substituents in

more complex systems (Merritt and Loening 1979).

Part I

7

Figure: 1.5 Fischer (left) and IUPAC (right) nomenclature systems for porphyrins

1.1.2.2 Synthetic Methodologies of Porphyrins

Fischer synthesized and named protoporphyrin-IX in 1929 for the first time. Afterwards,

a large number of synthetic routes have been developed for the synthesis of porphyrin

and its derivatives for structural, mechanistic, and biological studies. Although

porphyrins can be generated from the total synthesis from monopyrrolic subunits, but the

types of porphyrins that can be generated by this scheme are very limited. Recently,

many improved synthetic routes have been reported, which now provide easy access to

useful porphyrins. Now a days one of following methods can be employed for the

synthesis of porphyrin:

Rothemund Method

Alder-Longo Method

Lindsey Method

Mc Donald (2+2 synthesis) Method

Rothemund Method

Rothemund was the first scientist who introduced monopyrrole tetramerization to tetra-

arylporphyrin synthesis (Rothemund 1939). His method involved the heating of

Part I

8

benzaldehyde with pyrrole in pyridine as solvent in a sealed tube, to producemeso-

H2TPP, this reaction is called the Rothemund reaction. Zn (OAc)2 was used to act as

template in the presence of high pressure resulting 11% yield. Due to the high

temperature and pressure the reaction occurs in lower yields. When the reaction was

carried out in a mixture of methanol/pyridine at atmospheric pressure, both meso-H2TPP

and the chlorin (dihydro) form of meso-H2TPP were obtained. The chlorin is then

converted to meso-H2TPP by oxidization with oxygen or DDQ. This method has

applications with only a limited number of aldehydes.

Alder-Longo Method

Adler and Longo modified the Rothemund methodin 1967 (Adler, Longo et al.

1967).They studied many solvent systems along with a variety of salts to enhance the

formation of meso-H2TPP. The reaction conditions were relatively mild, from which a

much higher yield was achieved as well as the rate of reaction, was much faster as

compared to Rothemund method. In the Alder-Longo method refluxing pyrrole,

benzaldehyde in propionic acid at atmospheric pressure is used to affordthemeso-H2TPP

in 20% yield (Scheme 2.1). Although this method have a significant advantage over the

Rothemund method, but the Adler-Longo methodology have certain vexing problems,

which can be stated as:

The reaction conditions are relatively less selective towards some reactive

functionalities.

The high level of tar produced presents purification problems in mixed aldehyde

reaction mixture.

The reaction has poor reproducibility.

But despite all these disadvantages, it was still the most efficient method for the synthesis

of meso-tetra-alkylporphyrins at that time.

Part I

9

7

Scheme 1.1 Synthesis of meso-H2TPP (7) by Alder-Longo Method.

Lindsey Method

In 1986, Lindsey optimized the method with improved synthesis of porphyrins, under so

called Lindsey conditions. This is by far the most effective route for synthesizing

symmetrical porphyrins i.e. porphyrins with the same substituents on all four meso-

positions, or all eight of β-pyrrolic positions, and/or a combination of them. Lindsey

method is two-step process which consists of the formation of porphyrinogen from

monopyrrole tetramerization and then a separate oxidation (Scheme 1.2) (Lindsey, Hsu et

al. 1986).

Lindsey demonstrated that the formation of meso-H2TPP could be achieved under

equilibrium conditions, in which many functional groups could survive. A colorless

porphyrinogen was first formed, followed by a subsequent oxidation step with p-chloranil

or DDQ. meso-H2TPP was formed by dissolving benzaldehyde and pyrrole in

dichloromethane (DCM) in the presence of acid catalyst BF3•Et2O or TFA. The yields of

product under these conditions were improved to around 30-40 %. He further

concludedthat p-chloranil afforded higher yield as an oxidizing agent as compared to

DDQ.

Part I

10

7

Scheme 1.2 Synthesis of meso-H2TPP (7) by Lindsey Method.

McDonald (2+2 synthesis) Method

Finally, McDonald developed a series of linear steps for the formation of porphyrin with

greater degree of regioselectivityas compared to Lindsey and Alder methods. The

McDonald method involves condensation of two molecules of dipyrromethane one of

which bears substitutions at α-position while the other having no such substitution

(scheme 1.3). Model studies of porphyrins occurring in nature were carried out to study

ferrochelate enzyme by using modified MacDonald approach (Hoare and Heath 1960).

8

Scheme 1.3 Synthesis of meso-H2TPP by McDonald method.

Part I

11

1.1.2.3 Mechanism of Synthesis of Meso-H2TPP

7

Scheme 1.4 Proposed Mechanism for synthesis of meso-H2TPP

Formation of meso-H2TPP occursvia electrophilic substitution between pyrrole and

benzaldehyde in the presence of an acid. While the presence of phenyl rings in the

tetrapyrrole, open chain transition state increased the probability of cyclization as

opposed to aliphatic chain, due to the hindrance in rotation about the bridge carbon

atoms.

The condensation of pyrrole with benzaldehyde was carried out by dropwise addition of

pyrrole in high dilution to the aldehyde solution. Under these conditions, the

concentration of co-polmers, especially tetrapyrryl becomes considerably high and the tar

formation is lower.

Part I

12

1.1.3 Thiolated Cationic Heterocyclic Ligands for Stabilization of Metal Nanoparticles

Pyridinium compounds are known to form ionic liquids (ILs) which have been

extensively used as stabilizing/capping agents for metal nanoparticles (Dupont, Fonseca

et al. 2002; Kim, Demberelnyamba et al. 2003). Reports show that these systems usually

tend to cause aggregation of nanoparticles due to instability and their phase transfer

characteristics (Cassol, Umpierre et al. 2005; Wang and Yang 2005). The uses of

pyridinium ligands are not only limited to ILs but their wide spread occurrence in nature

make them fascinating candidate for biological and photophysical evaluation. Several

pyridinium nanoconjugates and complexes have recently being developed for catalysis

(Tourneux, Gornitzka et al. 2007; Zhu, Jang et al. 2011), drug delivery, biocidal activities

(Patil, Kokate et al. 2011) and molecular recognitions (Saha, Agasti et al. 2012;

Bagherzadeh, Pirmoradian et al. 2014).

Part I

13

1.2 RESULTS AND DISCUSSION

1.2.1 Synthesis and Applications of Polyfunctional Hydrogenated Pyridines from

Acetophenone Derivatives

Commercially available substituted acetophenone derivatives were converted into

alkylidene malononitrile via Knoevenagel condensation by following Barnes et al.

protocol. Towards our proposed synthetic target, we test the TBAF mediated cyclization

conditions to form dicyano substituted dihydropyridine derivatives (Scheme 1.5). The

desired products 9-26 were obtained in good to excellent yield (50% to 95%).

Scheme 1.5 Synthetic methodology of the target DHPs

The successful preparation of alkylidenemalononitriles in the first step enabled us to

explore their dimerization potential under solvent-free conditions for the preparation of

DHPs. The next step was the structure elucidation of these new dimerized compounds.

There are few possibilities in which the alkylidenemalononitrile could be dimerized to

give different positional isomeric structures as shown in Scheme 1.5.

The two interesting 1H NMR signals were observed in the spectra of different analogues

which proved to be very useful in solving the structures of these novel compounds, those

are, two doublets for two non-equivalent protons of methylene group around 3 ppm, and

a broad singlet for NH group around 6.5 ppm in chloroform. Furthurmore, compounds 9

Part I

14

and 11 were successfully crystallized with little effort in the Ethanol/H2O mixture, and

the X-ray crystallographic structures confirmed the correct assignment of NMR peaks.

The crystal structure clearly showed the attachment of malononitrile group to the C-2 via

double bond, cyano group at C-3 position, phenyl ring at C-4 position and quaternary

center at C-6 position (Fig. 1.6).

Figure: 1.6 X-rays crystallographic structures of compounds 9 and 11

All the spectroscopic data which includes 1H NMR, 13C NMR, FT-IR and high resolution

mass spectra showed complete agreement with the structure determination. The

dimerization of ortho substituted ylidenes (entry 14, 20, and 25 of table 1.4) was

unsuccessful which could be possibly explained in terms of steric hindrance.

A probable mechanism of this dimerization has been outlined in Scheme 1.6. The

carbanion, generated by the abstraction of proton from methyl group of alkylidene

malononitrile by TBAF, may rearrange itself to keteneimine intermediate, which may

then attack on dicycano alkene of another molecule to afford another reactive

intermediate carbanion. This intermediate then might undergo rearrangement to afford

the final product by cyclization.

Part I

15

Scheme 1.6 Plausible mechanism for the dimerization of alkylidene

malononitrile to give DHP

1.2.1.1 Urease Activity and Structural Activity Relationship

The urease inhibition activity analysis of the synthesized DHPs showed that most of the

derivatives are inactive in the series. However, entry 11 (IC50 = 20.4 ± 0.66 µM) showed

comparable urease inhibitory potential with standard inhibitor thiourea (IC50 = 21.0 ±

0.01 µM). While entry 12 (IC50 = 84.8 ± 6.99 µM), demonstrated moderate and entry 7

(IC50 = 244.6 ± 9.37 µM) showed weak inhibition potential (Table 1.1).

The 4,6-dimethoxy-5,6-dihydropyridine derivative demonstrated excellent inhibitory

potential among the series with IC50 values 20.4 ± 0.66, while 4,6-dihydoxy-5,6-

dihydropyridine derivative with an IC50 value 84.8 ± 6.99 µM exhibited moderate

activity. The difference between these structural analog’s activities suggests that change

in position and electronic effects of substituents on phenyl rings affect the urease

inhibitory activity.

Part I

16

Table 1.1 In vitro Urease activity of DHPs

Entry

Ring substituents IC50 (μM ± SEMA)

1 p-Br NAb

2 p-Cl NAb

3 acetophenone NAb

4 naphtha. NAb

5 p-I NAb

6 m-Br NAb

7 p-Me 244.66 ± 9.37

8 m-OMe NAb

9 p-F NAb

10 m-F NAb

11 p-OMe 20.48 ± 0.66

12 p-OH 84.87 ± 6.99

13 m-OH NAb

14 p-NO2 NAb

Thioureac 21.0 ± 0.01

SEMa is the standard error of the mean, NAb Not active

Thioureac standard inhibitor

It can be observed in table 1.1 that most of the electron withdrawing substituents and

meta position of electron donating groups in the series of these compounds are inactive.

The 4,6-dimethyl-5,6-dihydropyridine derivative with IC50 value 244.66 ± 0.01 µM also

exhibit weak inhibition due to low electron donating effects of para methyl substituent on

the phenyl rings of DHPs. Thus, it can be summarized that electron releasing groups at

para positions on phenyl ring greatly affect the urease inhibitory activity. This study

identifies a potent compound 11 (Table 1.1) which may serve as lead compound for

future research.

Part I

17

1.2.1.2 Anticancer Activity and Structural Activity Relationship

We also explored our DHP series against two cancer cell lines; PC3 and HeLa cell line.

The results of the anticancer activity (Table 1.2) on both cell lines showed that most of

the compounds are cytotoxic and have a varying degree of anticancer potential having

IC50 values ranging between 4.40 ± 0.12 to >30 µM, in comparison with standard

doxorubicin (IC50 value 0.91 ± 0.12 and 3.1 ± 0.20µM). Entry 1, and 2 showed highest

cytoxicity among the series in both cell lines as compared to doxorubicin. Entry 3, 4, 5, 6,

7, 8, 9, 10, and 11 demonstrated moderate cytoxicity, while compounds 12, 13, and 14

exhibited a weak activity towards both Cells line with IC50> 30.

The structure activity relationship can be constructed by closely observing the trend in

cytoxicity results. The variation in the anticancer activity through the series mainly

depends upon the position and electronic nature of the substituents on the phenyl rings.

The higher cytoxicities of p-Bromo and p-Chloro derivative, as compared to other

compounds in the series, can be understood on taking into account the electronic effect,

leaving group ability and reactivity of the substituents on the aromatic rings. The DHPs

with electron withdrawing groups having good leaving group character contained highest

cytotoxicity through the series. While the positions of the substituents also plays a pivotal

role in the anticancer activity, since the activity of 3-bromo derivative is lower than that

of 4-bromo derivative, whilst in the case of 3-methoxy derivative, activity is slightly

higher than 4-methoxy derivative. Moreover, an addition of phenyl ring as in the casse of

6-methoxynaphthalene dihydropyridine derivative also showed significant activity. These

results can be concluded as; electron withdrawing group having good leaving ability, as

well as their position on the phenyl ring significantly enhanced the anticancer activity of

the compounds. This study identifies compounds two potent novel anticancer agents

which may serve as lead compounds for the future research.

1.2.1.3 DPPH Radical Scavenging Activity

Free radical scavenging activity results show that all the DHP derivatives 1-13 are

moderately active to low active with a wide range of IC50 values from 127.4 ± 3.5 to

284.5 ± 0.66 (Table 1.3).

Part I

18

Table1.2Anticancer activity of DHPsagainst PC3 and HeLa Cell line

Entry Ring

substituents PC3 Cell line HeLa Cell line

IC50 (μM ± SEMA)

IC50 (μM ± SEMA)

1 p-Br 4.40 ± 0.13 8.81 ± 0.25

2 p-Cl 6.85 ± 0.34 8.81 ± 0.63

3 acetophenone 8.03 ± 0.22 23.06 ± 0.10

4 naphtha. 8.36 ± 0.48 14.67 ± 0.75

5 p-I 8.87 ± 0.59 9.67 ± 0.20

6 m-Br 9.62 ± 0.55 11.26 ± 0.85

7 p-Me 10.48 ± 0.24 18.84 ± 0.87

8 m-OMe 11.51 ± 0.45 20.18 ± 0.39

9 p-F 13.10 ± 0.62 23.09 ± 0.03

10 m-F 14.02 ± 0.42 26.41 ± 1.97

11 p-OMe 14.55 ± 0.38 14.73 ± 0.14

12 p-OH > 30 > 30

13 m-OH > 30 > 30

14 p-NO2 > 30 > 30

Doxorubicinb 0.91 ± 0.12 3.1 ± 0.20

SEMa is the standard error of the mean Doxorubicinb standard anticancer drug for many type of cancer cell lines

The derivatives including 3'-fluoro phenyl, 3'-bromo phenyl, 3'-methoxy phenyl, 4'-

bromo phenyl, 4'-iodo phenyl and 4'-methoxy phenyl exhibited moderate activity, while

the other derivatives in the series displayed low activity in comparison with standard

inhibitors ascorbic acid (IC50 = 40.6 ± 1.2μM) and butylated hydroxyanisole (IC50 = 40.6

± 1.2 μM).

Part I

19

Table 1.3 DPPH free radical scavenging activity

Entry

Substituted group (R) IC50 (μM ± SEM A)

1 phenyl NA b

2 4'-methyl phenyl 284.5 ± 0.66

3 4'-hydroxy phenyl 228.2 ± 3.36

4 3'-hydroxy phenyl 223.8 ± 3.30

5 4'-methoxy phenyl 161.4 ± 2.81

6 3'-methoxy phenyl 142.2 ± 0.60

7 4'-fluoro phenyl 172.8 ± 1.72

8 3'-fluoro phenyl 127.4 ± 3.5

9 4'-chloro phenyl 164.6 ± 4.50

10 4'-bromo phenyl 144.7 ± 2.46

11 3'-bromo phenyl 132.5 ± 3.32

12 4'-iodo phenyl 153.7 ± 0.50

13 6'-methoxynaphthalenyl 196.3 ± 6.5

14 Ascorbic acid c 40.6 ± 1.2

15 Butylated hydroxyanisole (BHA) c 44.7 ± 0.67

a SEM is the standard error of the mean. b NA: Not active.c Standard inhibitor for DPPH

radical scavenging activity.

Part I

20

1.2.2 Synthesis of Mannose Conjugated Porphyrin

1.2.2.1 Synthesis of Tetraphenylporphyrin (TPP) (7)

7

Scheme 1.7 Synthesis of meso-H2TPP (7)

TPP was synthesized by a modified Alder method. Propanoic acid was heated at 90 oC,

and a solution of (2 g, 18.86 mmoles) benzaldehyde dissolved in propanoic acid 2.3 mL

was added very slowly in hot propanoic acid. Then after continuous heating of the

solution at reflux temperature, pyrrole (18.86 mmoles) solution in propanoic acid (1.3 mL

in 2.3 mL) was added in above mixture dropwise for half hour. The reaction was refluxed

and monitored by TLC. After completion (about 1.5hrs) the reaction mixture was cooled.

The reaction mixture was worked up by extracting the crude porphyrin in 50 mL ethyl

acetate thrice from 50 mL, 2.5M NaOH aqueous solution.

Solvent was evaporated and solid product was passed through silica gel column using

ethyl acetate and hexane as mobile phase (3:1 respectively). The yield of pure product

was 1.2 g, 10 %.

Part I

21

1.2.2.2 Nitration of TPP (27)

27

Scheme 1.8 Synthesis of MNPP (27)

TPP was nitrated with the help of NaNO2 in TFA. By controlling the amount of TFA and

reaction time mono nitrated phenyl porphyrin (MNPP) was obtained as a major product

along with dinitro phenyl porphyrin (Kruper Jr, Chamberlin et al. 1989). To a solution of

TPP (160 mg, 0.26 mmoles) in 10 mL TFA was added (90 mg, 1.30 mmoles) NaNO2 and

reaction mixture was stirred for an hour. Reaction mixture was quenched with 15mL

water, extracted with 25 mL DCM thrice. The combined organic phases were evaporated

under reduced pressure affording brown residue. MNPP was purified through silica gel

column chromatography (DCM, Hexane 3:1 to 1:1) to get 50 mg in 30% yield.

Part I

22

Figure: 1.7 NMR spectrum of MNPP (27)

1.2.2.3 Mechanism of Nitration of Meso-H2TPP

A solution of TFA and sodium nitrite was used as source of nitronium ion for the

regiospecific nitration of meso-H2TPP. The steps involved in the formation of nitronium

ion from TFA and sodium nitrite can be given as,

Scheme 1.9 Mechanism of nitration of meso-H2TPP

Part I

23

The basic mechanism involved in the mentioned reaction is electrophilic aromatic

substitution of meso- substituted phenyl ring of the meso-H2TPP.

1.2.2.4 Reduction of nitro group of MNPP

Scheme 1.10 Reduction of nitro group of MNPP

MNPP (100 mg, 0.15 mmoles) was dissolved in 0.5 mL HCl. Tin dichloride (100 mg,

0.52 mmoles) was added in the solution and was heated at 60 oC for 2.5 hrs. Reaction was

monitored by TLC. After 2.5 hours, ammonium hydroxide was added to neutralize the

reaction mixture and extracted with 25 mL DCM thrice to get crude product. Purification

was carried out by Silica gel Column chromatography (DCM, Hexane 1:3 to 3:1). Yield

of monoamino phenyl porphyrin (MAPP 28) was 50 mg, 53 %.

Part I

24

Figure: 1.8 EI-MS spectrum of MAPP (28)

1.2.2.5 Mechanism of Nitro group of Reduction

The nitro group in MNPP was successfully reduced into amino group with the help of tin

(II) chloride hence yielding MAPP. Different solvents were screened for the reduction of

nitroporphyrins, and HCl was found to be the best one as it was acting as solvent and

source of reduction. The conversion of nitrporphyrin to aminoporphrin takes place

through free radical mechanism as shown in scheme 1.11.

Scheme 1.11 Proposed mechanism for reduction of nitro group

Part I

25

1.2.2.6 Conjugation of MAPP with Mannose

Scheme 1.12 Synthesis of target molecule (29)

Mannose (15 mg, 0.083 mmoles) was dissolved in 10 mL ethanol, MAPP (50 mg, 0.079

mmoles) was added in the solution. Few drops of acetic acid were added and reaction

mixture was refluxed overnight (Ojala, Ostman et al. 2000). Precipitates formed were filtered

and washed with 20 mL each DCM and MeOH separately. The residue was dissolved in

DMSO and was subjected to NMR for characterization. Yield was 47 mg, 75 %.

Figure: 1.9 NMR spectrum of mannose conjugated porphyrin (29)

Part I

26

1.2.2.7 Antibacterial Study of Mannose Conjugated Porphyrin via Photochemotherapy

Severe infections are produced including sight intimidating keratitis and deadly

granulomatous by pathogenic Acanthamoeba sp. (Marciano-Cabral and Cabral 2003;

Visvesvara, Moura et al. 2007). Regardless of the improvements made in supportive care

and antimicrobial chemotherapy to treat pathogenic Acanthamoeba, one of the most

upsetting aspects is that the death rate connected with GAE has remained significant

(90%). The existing diagnosis for the treatments of Acanthamoeba keratitis are expensive

and not totally operational against all strains (Pérez-Santonja, Kilvington et al. 2003), due

to the ability of the amoebae to form a strong cyst (Turner, Russell et al. 2004), which

results in the reappearance of infection. Therefore the treatment of pathogenic

Acanthamoeba keratitis is challenging. Complete knowledge about the pathophysiology

and pathogensis of infections associated with Acanthamoeba keratitis will certainly guide

to the improvement of diagnostic tools as well as therapeutic mediations (Clarke and

Niederkorn 2006). Recently it is recognized that a mannose binding protein (MBP) which

play a role in adhesion, as an exogenous mannose-block is expressed on the

Acanthamoeba membrane surface to adhere the parasites to microvascular endothelial-

cells of primary human brain and corneal epithelial cells (Garate, Cao et al. 2004).

Remarkable improvements were observed in Acanthamoeba sp. in Chinese hamster

model through oral immunization using recombinant mannose binding protein (Garate,

Cubillos et al. 2005).

Photochemotherapy is a relatively new technology that can treat only the target pathogens

and thus can be used as a therapeutic means. The basic principle of this technique is that

the photo-sensitizing compound is activated by the electromagnetic radiations which

results in generation of the nascent oxygen which induce cell necrosis in the target

pathogen (Hamblin and Hasan 2004; Huang, Dai et al. 2010). On the other hand, it is also

possible that host cells can bind with the photo sensitizing compound and then followed

by irradiation results in cell necrosis. Therefore to target a pathogenic Acanthamoeba sp.,

has persisted as a key concern (Baig, Iqbal et al. 2013). The main focus of our study is to

synthesize a photo-sensitizing compound such as porphyrin derivative conjugated with

mannose moiety to target specific Acanthamoeba and evaluate its in vitro potential. In the

Part I

27

current study mannose conjugated porphyrin derivative was prepared and subjected to

irradiation to kill the microbe.

Mannose conjugated porphyrin inhibited viability of A. castellanii

The A. castellanii trophozoiteswere incubated with two different concentrationof

mannose conjugated porphyrin i.e. 10 µM and 50 µM, to determine the amoebicidal

potential of mannose conjugated porphyrin on A. castellanii trophozoites. Which showed

that 50 µM concentration of mannose conjugated porphyrin revealed a significant

amoebicidal potential (P<0.001) as clearly visible in Fig. 1.10. Microscopic analysis also

confirmed the amoebicidal potential mannose conjugated porphyrin (Fig. 1.11). While A.

castellanii incubated with unconjugated mannose and unconjugated porphyrin having

concentration 50 µM did not showed any amoebicidal potential. The negative solvent

control groups also showed no effect on the A. castellanii tropozoites.

Figure: 1.10 Mannose-conjugated porphyrin (mann. por.) exhibited amoebicidal effects

against A. castellanii.

Part I

28

Figure: 1.11 A. castellanii was incubated with and without 50 µM mannose conjugatedporphyrin, and representative images were obtained at 100 x (A) and at400 x (B) magnification using a phase-contrast inverted microscope.

DMSO

Chloroform: Methanol

Mannose conjugated porphyrin

Mannose RPMI Alone

Porphyrin

Part I

29

Mannose conjugated porphyrin inhibited A. castellanii mediated host cell cytotoxicity

The mannose conjugated porphyrin was initially treated with A. castellanii, followed by

cytotoxicity assays of the host cells. The model group showed 95% 8.5% BMEC cell

deaths within 1440 minutes while the pretreated group with mannose conjugated

porphyrin significantly reduces the cytotoxicity of the host cells to 4.9% 2.5% (P <0.01)

as shown in Fig. 1.12A. A. castellanii was pretreated with mannose conjugated

porphyrin, which was followed by the performance of host cell cytotoxicity assays.

Without mannose conjugated porphyrin, A. castellanii produced 95% 8.5% BMEC cell

deaths within 24 h. The pretreatment of A. castellanii with mannose conjugated porphyrin

reduced host cell cytotoxicity significantly to 4.9% (P <0.01) compared to that with

amoebae alone (Fig. 1.12A). While unconjugated mannose, porphyrin, and solvents

showed no cytotoxicity of the host cells when they were separately incubated with A.

castellanii, and silmilarly solvent has showed no cytotoxicity to the microvascular

endothelial cell of the human brain (Fig. 1.12A). A coincubation assays was also

performed with microvascular endothelial cell of the human brain, along with mannose

conjugated porphyrin having concentration around 50 µM, and A. castellanii which also

reduces cytotoxicity of the host cells i.e. 58% 4% (P <0.01) as shown in Fig. 1.12B.

A

Part I

30

Figure: 1.12 (A). A. castellanii (5 x105) preincubated with and without 50 µM mannose-conjugated porphyrin (mann. por.) and exposed to light. (B). A. castellanii (5 x105) were coincubated with human BMEC and 50µM mannoseconjugatedporphyrin and exposed to light.

Mannose conjugated porphyrin blocked excystation in A. castellanii

The A. castellanii cysts were incubated in two medium which were capable of growth,

one A. castellanii cysts groups were initially treated with mannose conjugated porphyrin

while the other group was not treated to mannose conjugated porphyrin. The results

showed that the group A. castellanii cysts which were not treated with mannose

conjugated porphyrin appeared as workable trophozoites (1.2 x 106 amoebae) as shown in

Fig. 1.13. While the group treated with mannose conjugated porphyrin having 50 µM

concentration significantly repressed the A. castellanii excystation within three days (P

<0.01). The negative control groups with unconjugated mannose, porphyrin and solvents

showed sustainable trophozoites which were appeared in the well plates having A.

castellanii cysts after three days (Fig. 1.13).

B

Part I

31

Figure: 1.13 Mannose-conjugated porphyrin inhibited excystation of A. castellanii cysts. A.

castellanii cysts were scraped from nonnutrient agar plates, incubated with 50µM mannose porphyrin, and exposed to light.

Different drugs, like neomycin,chlorhexidine, propamidine isethionate, and

polyhexamethylene biguanide, are used to cure Acanthamoebainfections, but the recurrence

of these infections after a year or so is the main issueassociated with drugs. So therefore

complete treatments of diseases associated with Acanthamoeba toxicities is a challenging

work in the current scenario. Ficker et al. reported a fascinating study by showing that during

the treatments of Acanthamoeba keratitis, results in the resistance developments of

propamidine which ultimately led into the reappearance of infections. Different research

groups have shown that Acanthamoeba sp. resist to the antimicrobial treatments via

chemotherapy. In the current studies, we demonstrated the possible application of

photochemotherapy for the treatment of Acanthamoeba infections. (Aqeel, Siddiqui et al.

2015), a photo activated compound was synthesized from porphyrin and then it was

conjugated with mannose, in order to target the Acanthamoeba selectively. The mannose-

conjugated porphyrin showed potential applications in amoebicidal assays and can inhibited

the excystation effectively. The basic principle of this technique is that the photo sensitizing

compound is activated by the electromagnetic radiations which results in generation of the

Part I

32

nascent oxygen which induce cell necrosis in the target pathogen. The specific delivery of

photochemotherapeutic compounds to the target site is the considered to be the major

challenge. It was reported that mannose was considered to be the sugar with which

pathogenic Acanthamoeba can binds specifically in the presence of other sugars; MBP is

identified as a main adhesin which is expressed on the Acanthamoeba membrane surface.

Therefore, mannose was conjugated to photoactivated compound, a step towards targeted

drug delivery.

Importantly, the co-treatments and pretreatments of mannose conjugated porphyrin with A.

castellanii showed different results towards cytotoxicities of the host cells that is 58% and

4% respectively. A possible explanation of the higher cytotoxicity of the host cells in the co-

treatments of mannose conjugated porphyrin is the ROS generation by the mannose

conjugated porphyrin, which is a photosentising compound and in presence of brain cells of

human may have be involved to increases the cytotoxicity of the host cells. The mannose

conjugated porphyrin has also inhibited the excystation and complete inhibition of the

excystation assay was observed at 50 µM concentration of mannose-conjugated porphyrin.

This result was fascinating, as it was believed cysts don’t adhered to the host cells as MBP

was not expressed in the membrane surface, but here it seems that Acanthamoeba cysts

actually expresses the MBP on the membrane surface, which needs further experiments. The

lengthy exposure of photoactivated compound like mannose conjugated porphyrin and lipid

soluble compound were considered to be responsible for the complete inhibition of

excystation assays (Ferro, Coppellotti et al. 2006; Chen, Xuguang et al. 2008) which results

the reappearance of tropozoites from the cyst stage. The molecular mechanism associated

with aggregation of photo-dynamic compound at the Acanthamoeba cyst, and the movement

of this compound via operculum membrane to the trophozoites of Acanthamoeba remain to

be investigated in future along with its in vivo evaluation. In future porphyrin will be

conjugated to Acanthamoeba specific antibody for treatments of Acanthamoeba infections for

enhanced targeted therapeutic potential. Furthermore, pulsed interval treatments can also be

introduced, which will be more effective for clinical treatments. This type of treatment will

be very useful, as light used in this therapy is in the visible range which will never damage

tissues. The photodynamic therapy has been also useful for diverse diseases (Chen, Keltner et

al. 2002; Selbo, Kristian et al. 2002; Silva, Filipe et al. 2006) to kill Acanthamoeba in a less

harmful way, and it should be further investigated for in vivo applications.

Part I

33

1.3 EXPERIMENTAL

All the chemicals (Solvents and reagents) used were analytical grade and used as

purchased, without any pre-treatment until stated. 4-Pyridine carboxaldehyde, Pyrazine,

and Dimethyl aminopyridine were purchased from Tokyo chemicals industry (TCI,

ChinA). Dibromo propane and potassium thioacetate were purchased from Alfa-Aesar

(MA, USA). Instruments used for the analysis are, UV-visible spectrophotometer

Evolution 300, FT-IRspectra were recorded on Shimadzu FTIR 8900 spectrometer and 1HNMR spectra were recorded on Bruker 300 MHz and 400 MHz spectrometers.Mass

spectra were obtained on JEOL MS Route 600 H spectrometer at positive mode by using

electron impact (EI+) technique, HRMS was carried out at Thermo Finnigan MAT 95XP

instrument, while Matrix assisted laser desorption ionization (MALDI) mass

spectrometer Bruker Ultra flex, TOF-TOF. Coupling constant were calculated in Hertz

(Hz). The Merck silica gel 60 was used for column chromatography with commercially

available double distilled ethyl acetate, DCM and hexane as eluent.

1.3.1 General Procedure for the Synthesis of Polyfunctional DHP Derivatives

An oven dried screw-capped vial having magnetic stirrer bead was charged with

alkylidenemalononitrile (1.0 equiv) and neat TBAF.3H2O (1.0 equiv). The reaction

mixture was heated at 85-90 °C with continuous stirring for 15 h. The reaction progress

was monitored by TLC analysis (ethyl acetate hexane 1:4). The reaction mixture was

cooled to room temperature, diluted with 3 mL dioxane and then 100 mL, 1.0 M HCl

aqueous solution was added to remove excess TBAF. The resulting precipitates were

filtered and washed with more 1.0 M HCl to give the pure compounds in excellent yield

(52% to 98%). The structures of these compounds were characterized by using different

spectroscopic techniques including 1H NMR, 13C NMR, FT-IR, and EI-HRMS

spectroscopy.

Part I

34

Table 1.4 Structures and spectroscopic data of synthesized DHPs

S. No.

Structures with HR-

EIMS

And Melting Points

%

Yield

1HNMR

(Chemical shift in ppm)

FT-

IR1/cm

9

m/z = 336.1355 193-195 oC

98

7.50-7.40 (6H, m, ArH), 7.35 (2H app d, J = 8.0 Hz, ArH), 7.24 (2H app d, J = 8.0 Hz, ArH), 6.58 (1H, brs, NH), 3.49 (1H, d, J = 18.0 Hz, CH2), 3.15 (1H, d, J = 18.0 Hz, CH2), 1.79 (3H. s, CH3)

3448, 2925,2215,1598,1546,1445

,1257

10

m/z = 364.1645 125-128 oC

95

7.32 (2H, d, J = 8.4 Hz, ArH), 7.25-7.18 (4H, m, ArH), 6.11 (2H, d, J = 8.0 Hz, ArH), 6.50 (1H, br s, NH), 3.46 (1H, d, J

= 18, CH2), 3.09 (1H, d, J = 18, CH2), 2.37 (3H, s, CH3), 2.33 (3H, s, CH3), 1.76 (3H. s, CH3)

3391, 3279, 2923, 2214, 1591, 1427, 1259

11

m/z = 372.166 120-122 oC

75

7.39 (2H, app dd, J = 8.8 Hz, 5.0 Hz, ArH), 7.23 (2H, app dd, J = 9.0 Hz, 5.0 Hz, ArH), 7.12 (4H, app dd, J = 16 Hz, 8.5 Hz, ArH), 6.41 (1H, br s, NH), 3.40 (1H, d, J = 18 Hz, CH2), 3.15 (1H, d, J = 18 Hz, CH2), 1.79 (3H, s, CH3)

3446, 2216, 1602, 1507, 1432, 1237

12

m/z = 372.1176

130-135 oC

75

7.45-7.39 (2H, m, ArH), 7.22-7.15 (2H, m, ArH), 7.07 (1H, app td, J = 8.0 Hz, 2.0 Hz, ArH), 7.03-6.99 (2H, m, ArH), 6.94 (1H, app dt, 7.6, 2.0, ArH), 6.15 (1H, br s, NH), 3.40 (1H, d, J = 18 Hz, CH2), 3.16 (1H, d, J = 18 Hz, CH2), 1.79 (3H, s, CH3)

3249, 2222, 2204, 1614, 1584, 1487, 1437, 1270

Part I

35

13

m/z = 404.0571 132-135 oC

90

7.42 (4H, app t, J = 8.8 Hz, ArH), 7.22 (2H, d, J= 8.7 Hz, ArH), 7.16 (2H, d, J = 8.7 Hz, ArH), 6.43 (1H, br s, NH), 3.39 (1H, d, J = 18 Hz, CH2), 3.15 (1H, d, J = 18 Hz, CH2), 1.78 (3H. s, CH3)

3449, 2218, 1593, 1542, 1489, 1260

14

No React-ion

--- ---

15

m/z = 492.9691 138-140oC

90

.56 (4H, app dd, J = 16 Hz, 8.4 Hz, ArH), 7.21 (2H, d, J = 8.4 Hz, ArH), 7.09 (2H, d, J = 8.4 Hz, ArH), 6.83 (1H, br s, NH), 3.38 (1H, d, J = 18 Hz, CH2), 3.15 (1H, d, J = 18 Hz, CH2), 1.77 (3H, s, CH3)

3438, 1591, 1544, 1429, 1259

16

m/z = 492.9691

148-150 oC

84

7.63 (1H, app dt, J = 6.4 Hz, 1.6 Hz, ArH), 7.51 (1H, d, J = 7.6 Hz, ArH), 7.40 (2H, app d, J = 6.4 Hz, ArH), 7.34-7.30 (H, m, ArH), 7.16 (1H, d, J = 7.6 Hz, ArH), 6.15 (1H, br s, NH), 3.39 (1H, d, J = 18 Hz, CH2), 3.14 (1H, d, J = 18 Hz, CH2), 1.79 (3H, s, CH3)

3447, 3285, 2220, 2190, 1585, 1560, 1433, 1408

17

m/z = 587.9291

89

7.77 (4H, app dd, J = 17 Hz, 8.8 Hz, ArH), 7.07 (2H, d, J =

8.4 Hz, ArH), 6.96 (2H, d, J =

8.8 Hz, ArH), 6.41 (1H, br s, NH), 3.37 (1H, d, J = 18 Hz, CH2), 3.14 (1H, d, J = 18 Hz, CH2), 1.77 (3H. s, CH3)

3441, 1589, 1548, 1429, 1261

Part I

36

18

m/z = 368.1273

253-255 oC

75

10.55 (1H, br s, NH), 9.55 (1H, br s, ArOH), 9.45 (1H, br s, ArOH), 7.50 (2H, d, J = 8.7 Hz, ArH), 7.17 (2H, d, J = 8.4 Hz, ArH), 6.87 (2H, d, J = 8.7 Hz, ArH), 6.73 (2H, d, J = 8.4 Hz, ArH), 3.75 (1H, d, J= 18 Hz, CH2), 3.14 (1H, d, J= 18 Hz, CH2), 1.65 (3H, s, CH3).

3382, 1575, 1510, 1435, 1261

19

m/z = 368.1273 270-272 oC

93

9.92 (1H, br s, NH), 9.65 (1H, br s, ArOH), 9.55 (1H, br s, ArOH), 7.29 (1H, app t, J = 7.8 Hz, ArH), 7.18 (1H, app t, J = 7.8 Hz, ArH), 6.92 (1H, d, J = 8.0 Hz, ArH), 6.85-6.78 (3H, m, ArH)), 6.70-6.65 (2H, m, ArH), 3.61 (1H, d, J= 18 Hz, CH2), 3.27 (1H, d, J= 18 Hz, CH2), 1.67 (3H, s, CH3).

3449, 3250, 2213, 1594, 1451, 1429

20

No React-ion

--- ---

21

m/z = 396.1515 119-121 oC

95

7.45 (2H, d, J = 9.0 Hz, ArH), 7.15 (2H, d, J = 8.7 Hz, ArH), 6.91 (4H, app t, J = 9.4 Hz, ArH), 6.73 (1H, br s, NH), 3.82 (3H, s, OCH3), 3.78 (3H, s, OCH3), 3.46 (1H, d, J = 18 Hz, CH2), 3.05 (1H, d, J = 18 Hz, CH2), 1.75 (3H, s, CH3).

3449, 2926, 2212, 1510, 1459, 1259.

Part I

37

22

m/z = 397.1638

133-135 oC

80

7.34 (2H, app t, J = 8.0 Hz, ArH), 7.01 (1H, app dd, J = 8.0 Hz, 2.0 Hz, ArH), 6.93 (1H, d, J = 8.0 Hz, ArH), 6.87-6.79 (3H, m, ArH), 6.73 (1H, app t, J = 2.1 Hz, ArH), 6.38 (1H, br s, NH), 3.80 (3H, s, OCH3), 3.79 (3H, s, OCH3), 3.45 (1H, d, J = 18 Hz, CH2), 3.11 (1H, d, J = 18 Hz, CH2), 1.77 (3H, s, CH3)

3449, 3284, 2212, 1598, 1427

23

m/z = 427.1130

52

9.68 (1H, br s, NH), 8.30 (2H, dd, J = 9.0 Hz, ArH), 8.26 (2H, d, J = 9.0 Hz, ArH), 7.73 (2H d J = 9.0 Hz, ArH), 7.62 (2H, d, J = 9.0 Hz, ArH), 3.70 (1H, d, J = 18 Hz, CH2), 3.53 (1H, d, J

= 18 Hz, CH2), 1.77 (3H, s, CH3).

3447, 3419, 2963, 2928, 2188, 2160, 1596, 1520, 1453

24

m/z = 426.1068.

80

9.84 (1H, br s, NH), 8.37 (1H, app dt, J = 8.0 Hz, 2.0 Hz, ArH), 8.33 (1H, br s, ArH), 8.26 (2H, app d, J = 9.6 Hz, ArH), 8.20-8.13 (3H, m, ArH), 8.07 (1H, d, J = 8.0, Hz, ), 7.94-7.89 (2H, m, ArH), 7.84 (1H, app d, J = 3.6 Hz, ArH), 7.80 (1H, d, J = 8.0 Hz, ArH), 7.74 (1H, d, J = 8.0 Hz, ArH), 7.70-7.62 (3H, m, ArH), , 3.90 (1H, d, J = 18 Hz, CH2), 3.46 (1H, d, J = 18 Hz, CH2), 1.78 (3H, s, CH3)

3447, 3382, 3266, 2219, 2202, 1612, 1523, 1437

25

No reacti-on

--- ---

Part I

38

26

m/z = 496.1912

148-150 oC

65

7.90 (1H, app d, J = 1.4 Hz, ArH), 7.81 (1H, app d, J = 8.8 Hz, ArH), 7.81 (3H, app d, J =

8.8 Hz, ArH), 7.60 (1H, app d, J = 1.4 Hz, ArH), 7.43 (1H, dd, J = 8.8 Hz, 2.0 Hz, ArH), 7.38 (1H, dd, J = 8.8 Hz, 2.0 Hz, ArH), 7.19 (2H, app td, J

= 8.8 Hz, 2.8 Hz, ArH), 7.12 (1H, d, J = 2.0 Hz, ArH), 7.08 (6H, d, J = 2.4, Hz, ArH), 6.49 (1H, br s, NH), 3.91 (6H, s, 2 x OCH3), 3.70 (1H, d, J = 18 Hz, CH2), 3.27 (1H, d, J = 18 Hz, CH2), 1.89 (3H, s, CH3)

3448, 2926, 2212, 1627, 1579, 1540, 1482, 1269.

1.3.2 Urease Inhibition Assay (In vitro)

The reaction mixture is consist of the solutions of 25 µL jack bean urease enzyme, 55 µL

of its substrate having 100 mM urea. Reaction mixture was incubated with 5 µL of DHPs

(0.5 mM concentration) at 30 ºC for 15 min in 96-well plates. Then, the urease inhibitory

activity was evaluated by determining ammonia production via indophenol method

(Weatherburn 1967). Briefly, 45 µL of phenol reagent (1% w/v phenol with 0.005% w/v

sodium nitroprussside), and 70 µL of alkali reagent (0.5% w/v NaOH with 0.1% active

chloride NaOCl) were added to each well. The change in absorbance per minute was

measured after 55 min at 630 nm using a microplate reader (Molecular Device, USA). All

the reactions were performed in triplicate in a final volume of 200 µL and the results

(change in absorbance per min) were processed by using softMax Pro software

(molecular Device, USA). The pH was maintained at 6.8 in all assays, while the

percentage inhibitions were calculated from the formula 100-(ODtestwell/ODcontrol) ×100.

Thiourea was used as the standard inhibitor of urease (Table 1.1).

1.3.3 Anticancer Activity Assay (In vitro)

Cytotoxicities of compounds were evaluated in 96-well flat-bottomed micro plates via

standard MTT (3-[4, 5-dimethylthiazole-2-yl]-2, 5-diphenyl-tetrazolium bromide)

Part I

39

colorimetric assay (Mosmann 1983). PC3 and HeLa cells (Prostrate Cancer) were

cultured in Dulbecco’s Modified Eagle Medium, which was supplemented with 5% fetal

bovine serum (FBS), 100 IU/mL penicillin, 100 µg/mL streptomycin in 75 cm2 flasks,

and kept in 5% CO2 incubator at 37ºC. The exponentially growing cells were harvested,

counted by using haemocytometer, and then diluted with a particular medium. Cell

culture with the concentration of 1x105 cells/mL was prepared and 100 µL/well of this

solution was introduced into 96-well plates. After overnight incubation, medium was

removed and 200 µL of fresh medium was added with multiple concentrations of

compounds ranging in 5-50 µM. After 48 h, 200 µL MTT (0.5 mg/mL) was added to

each well and incubated for 4 more hours. Subsequently, 100 µL of DMSO was added to

each well. The extent of MTT reduction to formazan within cells was calculated on a

micro plate reader (Spectra Max plus, Molecular Devices, CA, USA) by measuring the

absorbance at 570 nm. The cytotoxicity was recorded as IC50 for both cell lines. The

percent inhibition was calculated by using the following formula: % inhibition = 100-

((mean of O.D of test compound – mean of O.D of negative control)/ (mean of O.D of

positive control – mean of O.D of negative control)*100). Doxorubicin was used as the

standard anticancer drug (Table 1.2).

1.3.4 DPPH (1,1-Diphenyl-2-Picryl Hydrazyl) Free Radical Scavenging Assay (In vitro)

The antioxidant activity of DHPs against 1,1-diphenyl-2-picrylhydrazil (DPPH) radical

was determined by following reported literature protocol.(Morales and Jiménez-Pérez

2001; Shaheen, Ahmad et al. 2005) The test sample (5μL, 1mM in DMSO) was added to

95 μL of DPPH (Sigma, 300μM) in ethanol and the resulting mixture was introduced into

a 96-well plate. The reaction mixture was incubated for half an hour at 37º C, and then

absorbance was determined at 515 nm on micro plate reader (Molecular Devices, USA).

DPPH radical scavenging activity was measured by comparing with a DMSO containing

control. All the assays were performed in dark and analysed in triplicate. The IC50 values

± SD of each compound were recorded and presented in the Table 1.3. Ascorbic acid and

Butylated hydroxyanisole (BHA) were used as standard inhibitors for DPPH radical

scavenging activity.

Part I

40

1.3.5 Acanthamoeba castellanii and Microvascular Endothelial Cells Cultures

All chemical reagents were obtained from Sigma laboratories England. A clinical isolate

of Acanthamoeba castellanii belongs to strain ATCC-50492. Amoebae were routinely

grown as reported in literature (Baig, Iqbal et al. 2013). The medium was every time

revitalized 16 to 21 hours before the start of experiments. The tropozoite forms of

amoebae were adhered to the walls of the flask and which were then collected and used in

all experiments. The microvascular endothelial cells were exclusively from human brain

which was isolated, cultured and purified as reported in literature (Stins, Gilles et al.

1997). The microvascular-endothelial cells of human brain were developed in collagen-

coated tissues of rat tail. The cytotoxicity assay was carried out by using microvascular-

endothelial cells of human brain which were grown in 24 well plates by 106 cells per mL

per well. The cell-density, confluent monolayers were molded within 1440 minutes and

were then used in subsequent experiments.

1.3.6 Synthesis of Mannose Conjugated Porphyrin

For the synthesis of mannose conjugated porphyrin, propionic acid (40 mL) was heated

up to 95 ºC followed by slow addition of benzaldehyde solution into hot propionic acid

which is prepared from 1.9 mL of benzaldehyde (18.85 mmoles) into 2.5 mL of propionic

acid. Then 1.3 mL of the pyrrole solution (18.85 mmmoles) was gradually added

dropwise in 30 minutes to the above solution at reflux temperature and was monitored by

TLC (DCM and MeOH 1:1). The reaction was completed in 1.5 h and then the reaction

mixture was cooled and the product tetraphenylporphyrin along with by products were

obtained through extraction in ethyl acetate (3 x 50 mL) from 50 mL of NaOH aqueous

solution having molarity 2.5 M. After solvent evaporation the solid residue were

subjected to silica column chromatography by using hexane and ethyl acetate in 9:1 ratio

respectively to get pure tetrahydroporphyrin in 7% yield. In the next step, 160 mg of

tetraphenylporphyrin (0.26 mmoles) was nitrated by continuous stirring for 1 h by adding

90 mg of NaNO2 having molarity 1.3 mM in 10 mL of TFA. Mono-nitrated

phenylporphyrin was obtained as a major product simply by controlling the reaction time

and TFA amount. The reaction was stopped by addition of water and then the product

Part I

41

was extracted in DCM (3 x 25 mL). After vacuum evaporation the pure mono nitrated

derivative was isolated in 30% yield by TLC with hexane and DCM with 1:1 to 1:3 ratio

respectively.

Then 100 mg of mono nitrophenylporphyrin (0.15 mmoles) along with 100 mg of tin

dichloride dehydrate (0.44 mmoles), were added to 0.5 mL of HCl and then the

temperature of reaction mixture was increased up to 60°C and continued for 2.5 h and the

progress of reaction was monitored by TLC. After reaction completion, the reaction

mixture was neutralized by addition of NH4OH and mono aminophenylporphyrin was

obtained in 53% yield after extraction with DCM (3 x 25 mL) and purified by TLC by

using hexane and DCM in 1: 3 to 3:1 ratio respectively. In the final step, 15 mg mannose

(0.079 mmoles) was mixed with 50 mg of mono aminophenylporphyrin (0.079 mmoles)

and the mixture was dissolved in ethanol (8 mL). Then, only three drops of glacial

CH3COOH were added and the reaction mixture was refluxed for 24 h. The product was

precipitated which was then filtered and washed with methanol and DCM separately. The

solid residue was dissolved in DMSO and was then subjected to NMR spectroscopy for

characterization and determination of the product purity. The synthesized product absorbs

electromagnetic radiation in the blue light range around 450 nm.

1.3.7Amoebicidal Assays

The amoebicidal assays were performed for mannose conjugated porphyrin, (12).

Mannose-conjugated porphyrin having molarity 10 µMand volume about 4 µl along with

mannose and porphyrin alone were incubated with A. castellaniifor 1 h in RPMI-1640

(106 amoebae per mL per well) in 24 well plates. After centrifugation the amoebae were

collected at 1,000 x g for about 5 min. After filtration the residue was re-suspended in 0.5

mL of RPMI-1640. Finally, the PYG was used for the re-suspension of amoeba pellet. In

the next step, lids were opened to expose to it to blue light inabiosafety hood for1hat

room temperature using a blue-round LED. Then these plates were incubated at 37°C for

24 h. Then hemocytometer was used for the amoeba counts and a phase contrast inverted-

microscope was used for imaging. The effect of solvents were checked by solvent

controls in which in same volume of DMSO i.e. 4-µl and 20-µl were used for amoebae

Part I

42

incubation, along with 50% chloroform and 50% methanol alone i.e. 1.34-µl and 6.7-µl

volumes of RPMI-1640. While in the positive controls, antiacanthamoebic drugchlorhexidine

was used at 20 µM in RPMI-1640 for incubation of the amoebae which showed 100%

killing of the amoebae.

1.3.8 Cytotoxicity assays of Host Cells

The mannose-conjugated porphyrin was subjected to in vitro antiamoebic assays by using

microvascular endothelial cells of the human brain. Mannose conjugated porphyrin were

incubated with A. castellanii for 1 h in RPMI-1640 (106 amoebae per mL per well) in 24-

well-plates and was exposed to light. After centrifugation the amoebae were collected at

1,000 x g for about 5 min. After filtration the residue was re-suspended in 0.5 mL of

RPMI-1640. Finally, the amoeba was incubated in 24 well plates in a 5% CO2 incubator

at 37 0C on the monolayers of microvascular endothelial cells of the human brain. On the

following day, a cell free accustomed medium was together and measuring of LDH

release give the host cells cytotoxicity assays by cytotoxicity detection kit. The

microvascular endothelial cells of the human brain were used a negative control while

monolayers lysed with 1% Triton X-100 were used as a positive control. The absorbance

value was converted to percent cytotoxicity by using (sample value negative value /high

value negative value) x 100 = % cytotoxicity.

In addition to the pretreatment of amoeba, the antiamoebic potential of mannose

conjugated porphyrin was also evaluated by coincubating microvascular endothelial cells

of the human brain with mannose-conjugated porphyrin and A. castellanii, as described

above. In the next step, the plates were incubated in a 5% CO2 incubator at 37 °C for 24

h, and cell free accustomed medium was gathered and by determining the LDH release,

the host cell cytotoxicity was evaluated.

1.3.9 Excystation Assays

Non-nutrient agar-plates were used for the excystation assays, which were prepared by

using distil water with 3% oxoid agar technical. Trophozoites of A. castellanii

(2x106amoebae) were immunized on the non-nutrient agar plates and accustomed for 15

Part I

43

days at 37 0C in order to allow the differentiation of tropozoites into cysts. No

trophozoites were then observed in phase-contrast inverted-microscope. The agar plates

were then swamped in 15 mL of PBS and keptat room temperature in a shaker for about

30 minutes. The cysts were then scratched off in a 50 mL tube and then were subjected

to centrifuge at 1,500 x g for about 10 minutes. The solid residue was re-suspended in 10

mL PBS. A hemocytometer was used to count the A. castellanii cysts. The excystation

assays were then carried out in order to evaluate the potential of mannose-conjugated

porphyrin on A. castellanii cysts (Lakhundi, Khan et al. 2014). Briefly, A. castellanii

cysts (5 x 105 cysts per mL per well) were accustomed with mannose conjugated

porphyrin having molarity 10 µM, along with mannose and porphyrin alone for about 1

h. The amoebae were gathered, washedwith PBS, and re-suspended in RPMI 1640 in 24

well plates. In the next step, lids were opened in order to exposed plates to

electromagnetic radiations for 1 h. In the final step, the amoebae cysts were gathered and

were then re-suspended in the growth medium (0.5 mL of PYG medium per well) in 24

well plates. The well plates were then incubated for three days at 37°C, which results in

the appearance of growing trophozoites under microscopic. A hemocytometer was used

to counts trophozoites.

1.3.10 Synthesis of thiolated heterocyclic ligands

Scheme 1.13 Synthesis of 1-(3-(acetylthio) propyl)-4-formylpyridinium (4-Py) (31)

Synthesis of 1-(3-bromopropyl)-4-formylpyridin-1-ium (30)

Pyridine carboxaldehyde (0.9 mL, 1 mmole) was stirred in dibromopropane (3 mL as

solvent) at 60 ºC for 8 hours. The brown precipitates formed after 08 hours which were

Part I

44

filtered and washed with 10 mL of methanol. The methanol filtrate was evaporated under

reduced pressure to get the N-alkylated product in viscous liquid state and was

characterized by Mass and NMR spectroscopy without any further purification. Yield 195

mg, 85 %. mp 140-145 ºC ESI-MS 227.96 M+. 1H NMR in MeOD, 9.04 (d, 2H, Ar,

metA), 8.16 (d, 2H, Ar, ortho), 4.89 (t, 2H, N-CH2), 3.52 (t, 2H, Br-CH2), 2.56 (m, 2H,

C-CH2-C). 13C NMR in MeOD, 161.7 (Ar, ipso), 146.3 (Ar, metA), 127.20 (Ar, ortho),

59.0 (CH2-N), 34.7 (CH2-Br), 29.6 (CH2-CH2-CH2).

Synthesis of 1-(3-(acetylthio)propyl)-4-formylpyridin-1-ium (31)

To a stirred solution of 1-(3-bromopropyl)-4-formylpyridin-1-ium (229 mg, 1 mmole) in

ethanol (8 mL; HPLC grade) at room temperature, potassium thioacetate (137 mg, 1.2

mmole) was added and the progress of reaction was monitored with thin layer

chromatography (TLC; dichloromethane: methanol 9:1). After 10 hours the solvent was

evaporated under reduced pressure to obtain brown solid crystalline residue which was

washed with methanol. The 1-(3-(acetylthio) propyl)-4-formylpyridin-1-ium (4-Py)was

recovered from filtrate. Yield 206 mg, 92%. mp 160-165 ºC ESI-MS 223.06 M+. 1H-

NMR in MeOD, 8.96 (d, 2H, Ar, metA), 8.33 (d, 2H, Ar, ortho), 4.65 (t, 2H, N-CH2),

2.92 (t, 2H, Br-CH2), 2.27 (m, 2H, C-CH2-C), 1.89 (s, 3H, CH3CO). 13C-NMR in MeOD,

166.4 (Ar, ipso), 145.1 (Ar, metA), 128.7 (Ar, ortho), 61.4 (CH2-N), 59.2 (CH2-S), 33.5

(CH3-CO), 22.1 (CH2-CH2-CH2).

Scheme 1.14 Synthesis of 1-(3-(acetylthio)propyl)pyrazin-1-ium (Pyr) (33)

Part I

45

Synthesis of 1-(3-bromopropyl)pyrazin-1-ium (32)

Pyrazine (160 mg, 2 mmol) was added in stirring 5 mL of dibromopropane (used as

solvent), and the reaction mixture was heated at 60 0C for 06 hours. The progress of

reaction was monitored with thin layer chromatography (TLC; dichloromethane (DCM):

methanol 9:1). After 06 hours, the excess dibromopropane was evaporated under vacuum

and 1-(3-bromopropyl)pyrazin-1-ium is obtained in viscous liquid state, which was used

without any further purification (378 mg, 94 %). MS (ESI-TOF): m/z: 200.9680 M+. 1H

NMR (300 MHz, MeOD): 9.50 (s, 2H), 9.15 (d, 2H), 4.88 (t, 2 H), 3.56 (t, 2 H), 2.63 (m,

2H). 13C NMR (75 MHz, MeOD): 152.50, 145.18, 62.14, 34.30, 29.04.

Synthesis of 1-(3-(acetylthio)propyl)pyrazin-1-ium (Pyr) (33)

To a stirred solution of 1-(3-bromopropyl)pyrazin-1-ium (202 mg, 1 mmole) in ethanol (8

mL; HPLC grade) at room temperature, potassium thioacetate (137 mg, 1.2 mmole) was

added and the progress of reaction was monitored by TLC (dichloromethane: methanol 9:1).

After 10 hours the solvent was evaporated under reduced pressure to obtain solid residue,

which was washed with DCM and recrystallized with 1:1 mixture of DCM and methanol to

afford 1-(3-(acetylthio)propyl)pyrazin-1-iumas brown crystalline solid (190 mg, 96%).

Melting point 278-282 oC. MS (ESI-TOF) m/z: 197.0686 M+. 1H-NMR (300 MHz, MeOD):

9.47 (s, 2H), 9.13 (s, 2H), 4.74 (t, 2 H), 2.94 (t, 2 H), 2.32 (s, 3H), 2.29 (m, 2H).

Scheme 1.15 Synthesis of ligand 1-(3-(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium (D2)

Part I

46

Synthesis of 1-(3-(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium (D2)(34)

Dimethyl aminopyridine (245 mg, 2 mmole), and Dibromomethane (1.6 mL, 16 mmole)

were stirred in 5 mL acetone at 60 oC for 30 minutes. Solvent was evaporated under

vaccou to obtain white precipitates of the mono substituted product 1-(3-bromopropyl)-4-

(dimethylamino) pyridin-1-ium. The precipitates (243 mg, 1 mmole) were dissolved in 5

mL ethanol and potassium thioacetate (140 mg, 1.2 mmole) was added. The reaction

mixture was stirred at 60 oC, while reaction progress was monitored by thin layer

chromatography (TLC; dichloromethane: methanol 9:1). After 10 hours the solvent was

evaporated under reduced pressure to obtain brown solid crystalline residue which were

filtered and washed with 10 mL of methanol. Methanol filterate was evaporated to get the

N-alkylated product (D2) in white crystalline state and was characterized by mass and

NMR spectroscopy. Yield 442 mg, 92%. Melting point 240-242 oC. ESI-MS 277.07

KM+. 1H-NMR in MeOD, 8.16 (d, 2H, Ar, metA), 6.99 (d, 2H, Ar, ortho), 4.21 (t, 2H, N-

CH2), 3.29 (s, 6H, N-CH3), 2.86 (t, 2H, S-CH2), 2.10 (m, 2H, C-CH2-C), 1.89 (s, 3H,

CH3CO). 13C-NMR in MeOD, 180.30 (Ar, ipso), 143.21 (Ar, metA), 109.01 (Ar, ortho),

57.21 (CH2-N), 40.34 (CH2-S), 32.13 (CH3-CO), 31.46 (CH3-N), 24.14 (CH2-CH2-CH2).

Part I

47

References

Abadi, A. H., T. M. Ibrahim, et al. (2009). "Design, synthesis and biological evaluation

of novel pyridine derivatives as anticancer agents and phosphodiesterase 3

inhibitors." Bioorganic & Medicinal Chemistry 17(16): 5974-5982.

Abdelrazek, F. M., N. A.-L. Kassab, et al. (2010). "Synthesis of some novel pyridine and

naphthyridine derivatives." European Journal of Chemistry 1(4): 368-372.

Adler, A. D., F. R. Longo, et al. (1967). "A simplified synthesis for meso-

tetraphenylporphine." The Journal of Organic Chemistry 32(2): 476-476.

Ajitha, M. J., S. Mohanlal, et al. (2012). "DPPH radical scavenging activity of tricin and

its conjugates isolated from "Njavara" rice bran: a density functional theory

study." Journal of Agricultral and Food Chemistry 60(14): 3693-3699 and

references therein.

Alden-Danforth, E., M. T. Scerba, et al. (2008). "Asymmetric cycloadditions of o-

quinone methides employing chiral ammonium fluoride precatalysts." Organic

letters 10(21): 4951-4953.

Amantini, D., R. Beleggia, et al. (2004). "TBAF-catalyzed synthesis of 5-substituted 1 H-

tetrazoles under solventless conditions." The Journal of Organic Chemistry 69(8):

2896-2898.

Aqeel, Y., R. Siddiqui, et al. (2015). "Photochemotherapeutic strategy against

Acanthamoeba infections." Antimicrobial Agents and Chemotherapy: AAC.

05126-05114.

Baig, A. M., J. Iqbal, et al. (2013). "In vitro efficacies of clinically available drugs against

growth and viability of an Acanthamoeba castellanii keratitis isolate belonging to

the T4 genotype." Antimicrobial Agents and Chemotherapy 57(8): 3561-3567.

Part I

48

Barnes, D. M., A. R. Haight, et al. (2006). "New conditions for the synthesis of

thiophenes via the Knoevenagel/Gewald reaction sequence. Application to the

synthesis of a multitargeted kinase inhibitor." Tetrahedron 62(49): 11311-11319.

Beckman, K. B. and B. N. Ames (1998). "The Free Radical Theory of Aging Matures."

Physiological Reviews 78(2): 547-581.

Bogdanowicz-Szwed, K. and M. Krasodomska (2006). "Synthesis of aryl-and pyridinyl-

substituted 2-amino-6-thioxopyridine-3-carbonitrile derivatives by tandem

Michael addition and cyclization reactions." Monatshefte für Chemie/Chemical

Monthly 137(3): 347-355.

Boucher, L. J. and J. J. Katz (1967). "Aggregation of metallochlorophylls." Journal of the

American Chemical Society 89(18): 4703-4708.

Burne, R. A. and Y.-Y. M. Chen (2000). "Bacterial ureases in infectious diseases."

Microbes and Infection 2(5): 533-542.

Chen, J., L. Keltner, et al. (2002). "New technology for deep light distribution in tissue

for phototherapy." The Cancer Journal 8(2): 154-163.

Chen, Z., S. Xuguang, et al. (2008). "In vitro amoebacidal activity of photodynamic

therapy on Acanthamoeba." British Journal of Ophthalmology 92(9): 1283-1286.

Chung, K., B. Moon, et al. (2006). "Oxidation of aromatic aldehydes with

tetrabutylammonium fluoride: Competition with the cannizzaro reaction."

Bulletin of Korean Chemical Society 27(8): 1203.

Clarke, D. W. and J. Y. Niederkorn (2006). "The pathophysiology of Acanthamoeba

keratitis." Trends in Parasitology22(4): 175-180.

Corey, E. and B. B. Snider (1972). "Total synthesis of (+-)-fumagillin." Journal of the

American Chemical Society 94(7): 2549-2550.

Part I

49

Cox, D. P., J. Terpinski, et al. (1984). "" Anhydrous" tetrabutylammonium fluoride: a

mild but highly efficient source of nucleophilic fluoride ion." The Journal of

Organic Chemistry 49(17): 3216-3219.

Cui, H., Y. Kong, et al. (2012). "Oxidative Stress, Mitochondrial Dysfunction, and

Aging." Journal of Signal Transduction 2012.

Dunkel, S., U. Hess, et al. (1997). "Synthese von cyanosubstiuierten Di‐und

Tetrahydropyridinen in DIMCARB (Dimethylamin–CO2‐Additionsverbindung)."

Journal für Praktische Chemie/Chemiker‐Zeitung 339(1): 414-419.

Ferro, S., O. Coppellotti, et al. (2006). "Photosensitized inactivation of Acanthamoeba

palestinensis in the cystic stage." Journal of Applied Microbiology 101(1): 206-212.

Garate, M., Z. Cao, et al. (2004). "Cloning and characterization of a novel mannose-

binding protein of Acanthamoeba." Journal of Biological Chemistry 279(28):

29849-29856.

Garate, M., I. Cubillos, et al. (2005). "Biochemical characterization and functional

studies of Acanthamoeba mannose-binding protein." Infection and Immunity

73(9): 5775-5781.

Hamblin, M. R. and T. Hasan (2004). "Photodynamic therapy: a new antimicrobial

approach to infectious disease?" Photochemical & Photobiological Sciences 3(5):

436-450.

Hargreaves, S. L., B. L. Pilkington, et al. (2000). "The synthesis of substituted

pyridylpyrimidine fungicides using palladium-catalysed cross-coupling

reactions." Tetrahedron Letters 41(10): 1653-1656.

Hiroya, K., R. Jouka, et al. (2001). "Cyclization reactions of 2-alkynylbenzyl alcohol and

2-alkynylbenzylamine derivatives promoted by tetrabutylammonium fluoride."

Tetrahedron 57(48): 9697-9710.

Part I

50

Hoare, D. and H. Heath (1960). "Dipyrromethanes and the biosynthesis of porphyrins."

Biochimica et Biophysica Acta 39(1): 167-169.

Hodgkin, D. C. (1979). New and Old Problem in the Structure Analysis of Vitamin B12.

Vitamin B12, Proceedings of the Third European Symposium on Vitamin B12

and Intrinsic Factors, Walter de Gruyter: Berlin, Germany.

Holiman, P. C. H., M. G. L. Hertog, et al. (1996). "Analysis and health effects of

flavonoids." Food Chemistry 57(1): 43-46.

Huang, L., T. Dai, et al. (2010). Antimicrobial photodynamic inactivation and

photodynamic therapy for infections. Photodynamic Therapy, Springer: 155-173.

Hunt, E. J., C. E. Lester, et al. (2001). "Effect of St. John's wort on free radical

production." Life Sciences 69(2): 181-190.

Johnson, D. S. and J. J. Li (2007). The Art of Drug Synthesis. New Jersey, John Wiley &

Sons, Inc.

Knoevenagel, E. (1898). "Condensation von Malonsäure mit aromatischen Aldehyden

durch Ammoniak und Amine." Berichte der Deutschen Chemischen Gesellschaft

31(3): 2596-2619.

Kruper Jr, W. J., T. A. Chamberlin, et al. (1989). "Regiospecific aryl nitration of meso-

substituted tetraarylporphyrins: a simple route to bifunctional porphyrins." The

Journal of Organic Chemistry 54(11): 2753-2756.

Lakhundi, S., N. A. Khan, et al. (2014). "Inefficacy of marketed contact lens disinfection

solutions against keratitis-causing Acanthamoeba castellanii belonging to the T4

genotype." Experimental Parasitology 141: 122-128.

Lemberg, R. and J. Barrett (1973). "Cytochromes." Journal of Cell Science 13: 1.

Part I

51

Lichitsky, B., A. Dudinov, et al. (2001). "Reaction of 3-aminocyclohex-2-en-1-ones with

arylidenemalononitriles: synthesis of N-substituted 1,4,5,6,7,8-hexahydro-

quinolin-5-ones."Arkivoc 9: 73-79.

Lindsey, J. S., H. C. Hsu, et al. (1986). "Synthesis of tetraphenylporphyrins under very

mild conditions." Tetrahedron Letters 27(41): 4969-4970.

Malicorne, G., J. Bompart, et al. (1991). "Synthèse et activité anti-bactérienne d'acides 4,

7-dihydro-4-éthyl-7-oxothiéno (3, 2-B) pyridine-6-carboxyliques." European

Journal of Medicinal Chemistry 26(1): 3-11.

Marciano-Cabral, F. and G. Cabral (2003). "Acanthamoeba spp. as agents of disease in

humans." Clinical Microbiology Reviews 16(2): 273-307.

Martínez-Martínez, F., R. Razo-Hernández, et al. (2012). "Synthesis and in vitro

Antioxidant Activity Evaluation of 3-Carboxycoumarin Derivatives and QSAR

Study of Their DPPH• Radical Scavenging Activity." Molecules 17(12): 14882-

14898 and references therein.

Menna, P., O. Gonzalez Paz, et al. (2012). "Anthracycline cardiotoxicity." Expert

Opinion on Drug Safety11(S1): S21-S36.

Merritt, J. and K. Loening (1979). "Nomenclature of tetrapyrroles." Pure and Applied

Chemistry 51: 2251-2304.

Meyer, A. S., M. Heinonen, et al. (1998). "Antioxidant interactions of catechin, cyanidin,

caffeic acid, quercetin, and ellagic acid on human LDL oxidation." Food

Chemistry 61(1–2): 71-75.

Milgrom, L. R. (1997). The Colours of Life: An Introduction to the Chemistry of

Porphyrins and Related Compounds, Oxford University Press Oxford.

Mobley, H., M. D. Island, et al. (1995). "Molecular biology of microbial ureases."

Microbiological Reviews 59(3): 451-480.

Part I

52

Morales, F. J. and S. Jiménez-Pérez (2001). "Free radical scavenging capacity of Maillard

reaction products as related to colour and fluorescence." Food Chemistry 72(1):

119-125.

Mosmann, T. (1983). Journal of Immunological Methods 65: 55-63.

Moure, A., J. M. Cruz, et al. (2001). "Natural antioxidants from residual sources." Food

Chemistry 72(2): 145-171.

Ojala, W. H., J. M. Ostman, et al. (2000). "Schiff bases or glycosylamines: crystal and

molecular structures of four derivatives of d-mannose." Carbohydrate Research

326(2): 104-112.

Pérez-Santonja, J. J., S. Kilvington, et al. (2003). "Persistently culture positive

Acanthamoeba keratitis: in vivo resistance and in vitro sensitivity."

Ophthalmology 110(8): 1593-1600.

Rice-Evans, C. A., N. J. Miller, et al. (1996). "Structure-antioxidant activity relationships

of flavonoids and phenolic acids." Free Radical Biology and Medicine 20(7): 933-

956.

Rothemund, P. (1939). "Porphyrin Studies. III. 1 The Structure of the Porphine2 Ring

System." Journal of the American Chemical Society 61(10): 2912-2915.

Selbo, P., a. l. Kristian, et al. (2002). "Photochemical internalisation: a novel drug

delivery system." Tumor Biology 23(2): 103-112.

Shaheen, F., M. Ahmad, et al. (2005). "Alkaloids of Aconitum laeve and their anti-

inflammatory, antioxidant and tyrosinase inhibition activities." Phytochemistry 66

(8): 935-940.

Silva, J. N., P. Filipe, et al. (2006). "Photodynamic therapies: principles and present

medical applications." Bio-medical Materials and Engineering 16: S147-S154.

Part I

53

Smith, K. M. and J. E. Falk (1975). Porphyrins and Metalloporphyrins, Elsevier

Amsterdam.

Stins, M. F., F. Gilles, et al. (1997). "Selective expression of adhesion molecules on

human brain microvascular endothelial cells." Journal of Neuroimmunology

76(1): 81-90.

Szökő, É. and T. Tábi (2010). "Analysis of biological samples by capillary

electrophoresis with laser induced fluorescence detection." Journal of

Pharmaceutical and Biomedical Analysis 53(5): 1180-1192.

Talelli, M., K. Morita, et al. (2011). "Synthesis and characterization of biodegradable and

thermosensitive polymeric micelles with covalently bound doxorubicin-

glucuronide prodrug via click chemistry." Bioconjugate Chemistry22(12): 2519-

2530.

Toyota, K., H. Shinkai, et al. (1993). Anticarcinogenic agents; potentiation, Google

Patents.

Turner, N., A. Russell, et al. (2004). "Resistance, biguanide sorption and

biguanide‐induced pentose leakage during encystment of Acanthamoeba

castellanii." Journal of Applied Microbiology 96(6): 1287-1295.

Villaño, D., M. S. Fernández-Pachón, et al. (2007). "Radical scavenging ability of

polyphenolic compounds towards DPPH free radical." Talanta71(1): 230-235.

Visvesvara, G. S., H. Moura, et al. (2007). "Pathogenic and opportunistic free‐living

amoebae: Acanthamoeba spp., Balamuthia mandrillaris, Naegleria fowleri, and

Sappinia diploidea." FEMS Immunology & Medical Microbiology 50(1): 1-26.

Volm, M., J. Mattern, et al. (1991). "Overexpression of P-glycoprotein and glutathione S-

transferase-pi in resistant non-small cell lung carcinomas of smokers." British

Journal of Cancer 64(4): 700.

Part I

54

Weatherburn, M. W. (1967). Analytical Chemistry 39: 971.

Williams, R., E. Spencer, et al. (2004). "Flavonoids: antioxidants or signalling

molecules?" Free Radical Biology and Medicine 36(7): 838-849.

Willstatter, R. and A. Stoll (1928). "Investigations on Chlorophyll: Methods and

Results."

Yasuhara, A., N. Suzuki, et al. (2002). "A concise synthesis of furostifoline by

tetrabutylammonium fluoride-promoted indole ring formation." Chemical and

Pharmaceutical Bulletin-Tokyo 50(1): 143-145.

Part II

55

CHAPTER 2

SYNTHESIS OF METAL NANOPARTICLES AND THEIR

APPLICATIONS

2.1 INTRODUCTION

2.1.1 Introduction of Nanotechnology

The concept behind nanoscience and nanotechnology started with a talk entitled “There’s

Plenty of Room at the Bottom” by physicist Richard Feynman at an American Physical

Society meeting held on December 29, 1959 at the California Institute of Technology

(CalTech). Feynman described the idea to manipulate and control individual atoms and

molecules. Over a decade later, Professor Norio Taniguchi coined the term

nanotechnology in his explorations of ultra-precision machining. Later on, with the

development of the atomic and electron microscopes in 1980s the modern

nanotechnology began.

Nanotechnology is emerging as a broad field in almost all field of science these days

especially in chemistry. It has the ability to produce materials, devices, and systems

having new highly controlled and fascinating properties by functioning at the atomic as

well as molecular levels. It utilizes exceptional physical and chemical properties that

appear when matter is constructed at the nanoscale (Blonder 2010).

The nanomaterials are distributed in vast variety according to their properties and

applications.Interface and colloid science has given rise to many materials which are

being usefully studied in nanotechnology, such as fullerenes, carbon nanotubes,

nanoparticles and nanoclusters.

2.1.2 Fullerenes

Fullerenes are molecules composed of carbons in the form of a hollow sphereand many

other shapes. Fullerenes are graphite analogues, which are composed of piled graphene

sheets of linked hexagonal rings, but the difference between graphene and fullerenes is

that fullerenes may also contain pentagonal or sometimes heptagonal rings. The first

Part II

56

fullerene, buckminsterfullerene or buckyball C60, was discovered in 1985 by Richard

Smalley, Robert Curl, James Heath, Sean O'Brien, and Harold Kroto. The encounter of

fullerenes greatly stretched the number of known carbon allotropes, which were limited

to graphite, diamond, and amorphous carbon. Buckyballs and buckytubes have been the

subject of concentrated research, both for their unique chemistry and for their

applications in materials science, electronics, and nanotechnology. The other known

natural fullerenes are C70, C76, C82 and C84 molecules, which are formed by lightning

discharges in the atmosphere from soot in trace amounts.

Figure: 2.1 Structure of a Fullerene

2.1.3 Carbon Nanotubes

Carbon nanotubes (CNTs) are carbon allotropes having a cylindrical nanostructure. The

graphite layer appears somewhat like a rolled-up chicken wire with a continuous

unbroken hexagonal mesh and carbon molecules at the apexes of the hexagons. Carbon

Nanotubes have several structures, which differ in length, thickness, and in the type of

number of layers. There are two main types of CNTs those are simply based on number

of walls, single walled carbon nanotubes (SWCNT) and multi walled carbon nanotubes

(MWCNT). Although they are formed from essentially the same graphite sheet, their

electrical characteristics differ depending on these variations, acting either as metals or as

semiconductors. As a group, Carbon Nanotubes typically have diameters ranging from <1

nm up to 50 nm. Individual nanotubes naturally align themselves into "ropes" held

together by van der Waals forces, more specifically, pi-stacking. These supramolecular

Part II

57

forces and unique structural features of CNTs make them ideally strong and excellent

conductor of both charge and heat.

Figure: 2.2 Depiction of CNTs

The recent literature is extremely rich with the examples and applications of CNTs. For

example, the H-bonding ability of cyclic peptides makes them striking candidate for the

formation of supramolecular nanotubes. Peptide nanotubes exist as tubular beta-sheet-like

structures which are formed by the self-association of flat, ring-shaped peptide subunits

made up of alternating D- and L-amino acid precursors. Peptide self-assembly formed by

extensive network of intersubunit hydrogen bonds (Ghadiri, Granja et al. 1993; Kim, Suh

et al. 2002).In another example, Calix[4]hydroquinone also self-assemble by H-bonding

and form nanotubes. In additon to hydrogen bonding, aromatic rings in Calix[4]

hydroquinone also displayed pi-pi stacking(Semetey, Didierjean et al. 2002).

2.1.4 Nanoparticles

Nanoparticles are nanodimensional materials (in the 1–100 nm size domains) which act

as a bridge between atomic and bulk materials and have been shown to exhibit a variety

of unique chemical, physical and electronic properties (Matsukawa, Watase et al. 2006).

As nanoparticles are essentially finely divided bulk materials, they are typically

thermodynamically unstable with respect to agglomeration. Consequently, they need to

Part II

58

be kinetically stabilized and this is typically done using a protective stabilizer. The

stabilization is achieved by electrostatic or steric forces or a combination of the two

known as “electrosteric forces”. The stabiliser is typically introduced during the

formation of the nanoparticles, the stabilizer may be organic ligands, surfactants,

polymers and dendrimers (Cookson 2012).

These NPs can be composed of a variety of materials including noble metals e.g. Au

(Boisselier and Astruc 2009) , Ag (Lu, Tung et al. 2008) (Podsiadlo, Paternel et al. 2005)

, Pd (Ung, Tung et al. 2009), semiconductors e.g. CdSe, CdS, ZnS (Boisselier and Astruc

2009) (Jamieson, Bakhshi et al. 2007), TiO2 (Drbohlavova, Adam et al. 2009), PbS

(Rogach, Eychmüller et al. 2007), InP (Rogach, Eychmüller et al. 2007), Si (O'Farrell,

Houlton et al. 2006). Metal nanoparticles can be synthesized by use of reducing agent

(Sun and Zeng 2002). Nanoparticles can be synthesized both in organic and aqueous

solution. Nano-structure materials including Au (Turkevich, Stevenson et al. 1951)

(Wang, Sato et al. 2003), Ag (Podsiadlo, Paternel et al. 2005), Co(Lu, Tung et al. 2008),

Nix(Verma, Giri et al. 2010), Fe2O3, FeO (OH) (Gnanaprakash, Mahadevan et al. 2007) ,

and CdTe (Bao, Wang et al. 2006) (Rogach 2000) have been synthesised in aqueous

solution. NPs composed of noble metals, transition metals, oxides and semiconducting

materials (Lu, Tung et al. 2008) (Brust, Walker et al. 1994) (Gittins and Caruso 2001)

(Sun, Murray et al. 2000) (Murray, Norris et al. 1993) have been synthesized in organic

solvents. Whilst much research has focussed on nanomaterials of the coinage metals

(especially those of gold) (Daniel and Astruc 2004). The most important of these metal

based nanoparticles are of noble metals such as gold and silver due to their

biocompatibility and photophysical properties.

Gold is the subject of interest in science from long time ago. Several gold catalysts and

compounds are formed in recent years that entertain chemistry to a variety of uses.

Recently a trend of gold compounds formation and their applications is developed in the

context of emerging nanosciences and nanotechnology with both nanoparticles and self-

assembled monolayers (SAMS) (Daniel and Astruc 2003). Gold nanoparticles are formed

by reduction of gold salts, conventionally of gold (III) derivatives. Turkevitch in 1951

reduced HAuCl4 in water using citrate (Turkevich, Stevenson et al. 1951). It leads to

Part II

59

AuNPs of size 20 nm. Size controlled methods consist of maintaining the ratio between

reducing/stabilizing agents (Frens 1973). One of the methods for gold nanoparticles

production is the Brust-Schiffrin two phase synthesis, the ratio between thiol/gold

determines the average size of forming nanospheres again. Larger ratios along with faster

addition of the reducing agent in cooled solutions give smaller Nanospheres (Brust,

Walker et al. 1994). AuNPs stabilized with thiols are of significant current interst due to

their ease in formation and substantial stabilities (Hutchings, Brust et al. 2008). Jadzinsky

et al., studied the structural features of AuNPs by X-Rays diffraction (Ackerson,

Jadzinsky et al. 2005; Jadzinsky, Calero et al. 2007).

Silver is another metal that has been widely used for nanoparticles synthesis since it is

relatively cheaper than the gold. Silver nanoparticles have been synthesized by a range of

different physical procedures such as evaporation/condensation technique (Kruis et. al

2000) and by using laser (Mafune et. al 2001), while chemical methods involve the

reduction of silver salts (Tao et. al 2006., Wiley et. al 2005).

Other metals based nanoparticles include zinc(Hattori, Mukasa et al. 2011), palladium

(Corthey, Rubert et al. 2012) etc. Metal oxide nanoparticles are also an important

paradigm in the nanotechnology; common examples are Fe (III) oxide (Rosen, Chan et al.

2012), titanium oxide (Shiraishi, Ikeda et al. 2011), zinc oxide etc. Some rare earth doped

nanoparticles are also reported (Bouzigues, Gacoin et al. 2011).

2.1.5 Nanoclusters

A group of atoms when reaches a size just too big to be called a molecule and too small

to show characteristic bulk properties and ranges between sub nm to 10 nm is termed a

nanocluster. Ideally the number of atoms is less than 100. Nanoclusters exhibit a

completely different behaviour from that predicted by scaling. There unique geometric

structure and quantized electronic states give them an exclusive set of properties. The

increase in number of atoms and consequent size play a vital role in controlling other

parameters responsible for the behaviour of these clusters. Individual clusters can be

assembled to build sub units for novel functional materials. The behaviour of

Part II

60

nanoclusters can be individually studied only when they are protected by a ligand to that

separated each cluster to avoid coalescence.

2.1.6 Synthesis of Nanoparticles and Nanoclusters

There are two main approaches which are most commonly employed to synthesize the

nanomaterials.

2.1.6.1 Bottom-up Approach

In this approach some chemical modifications utilized to arrange the smaller molecules

into complex molecular assemblies. These molecular self-assemblies basically operate on

the basic supramolecular interactions mainly so that a single molecule may arrange into a

useful conformations for the formation of larger assemblies.

2.1.6.2 Top-down Approach

Top-down approach is based on getting larger components to construct self-assembled

smaller molecular assemblies. In this methodology, physical methods for example laser

ablation etc. are mainly utilized to obtain the nanomaterials.

2.1.7 Dispersity in Nanoscale Synthesis

The vast applications and straightforward synthesis of nanostructures has made such a

fast growing area of research and synthesis is carried on every scale in every field.

Although the synthesis is simple there exist some limitations when it comes to size

selectivity and precision. Clusters of a limited size distribution and controlled nuclearity

have been obtained through different methods of separation as well as target oriented

synthesis but a technique that could yield monodisperse nanoclusters in bulk quantities is

yet to be devised.

2.1.8 Applications of Nanoparticles

Nanoparticles are explored in a variety of ways like their assessment of the in vivo

toxicity is also studied (Chen, Hung et al. 2009). Their use in in vitro assays, in vitro and

Part II

61

in vivo imaging, cancer therapy, and drug delivery are the hot topics in research these

days (Chen, Huang et al. 2008). Gold nanoparticles are very efficient drug delivery

systems because gold can easily be fabricated(Liu, Zhang et al. 1998). Other properties of

different metal based nanoparticles are surface Plasmon band (SPB) (Craig F. Bohren

1983), Fluorescent biological labelling (Bruchez, Moronne et al. 1998), Electrochemistry

(Miles and Murray 2003), DDSs (Mah C 2000), used for probing of DNA structures

(Mahtab, Rogers et al. 1995), used in hyperthermia (Shinkai, Yanase et al. 1999), used as

MRI contrast enhancers (Weissleder, Elizondo et al. 1990) etc. While their applications

in catalysis, supramolecular chemistry (Tshikhudo, Demuru et al. 2005; Ohyama, Hitomi

et al. 2009), molecular recognition (Xiao, Gao et al. 2012), biomarker detectors (Hong,

Huh et al. 2012), as sensors for example biosensors in food analysis (Pérez-López and

Merkoçi 2011), gas sensing (Sun, Liu et al. 2012), environmental applications include the

heavy metals removal from water (Hua, Zhang et al. 2012), automobile catalytic

convertor (Zhang, Cattrall et al. 2011), and their remarkable use in clinical diagnostic

(Larguinho and Baptista 2012).

2.1.9 Nanoclusters in Biomedical Sciences

2.1.9.1 Biological Imaging

Flourescence imaging technique offers certain advantages over other imaging techniques

like sensitivity and cost. Flourescent metal nanoclusters are excellent flourophores for

biological imaging due to their remarkably biocompatibile size and bright emission.

Human mesenchymal stem cells have been imaged by a 2 photon imaging technique

using dextran encapsulated Au nanoclusters as flourophores.

In vivo fluorescence imaging has also been reported in animals with bovine serum

albumen stabilized ultra-small Au clusters. High stability and low toxicity of the Au

nanoclusters have qualified them for use in continuous imaging. The nanoclusters were

intravenously injected into the mice then subjected to fluorescence imaging. The

fluorescent nanoclusters were soon visibly accumulated in the tumour sights where

permeability and retention have been enhanced. The nanoclusters stay in the living body

for 24 hours and afterwards are easily excreted through kidney filtration.

Part II

62

2.1.9.2 Biodetection

Good solubility in water, high emission, low toxicity are some of the reasons that make

nanoclusters so suitable for biodetection of different analytes in living systems.

Detection of Nucleic Acids

Among the other factors scaffolding is a main factor that effect the synthesis and

properties of nanoclusters. The emission properties of DNA-Ag nanoclusters have been

studied. As the nucleotide sequence changes, the fluorescence properties of nanoclusters

has also been changed this has led to the development of a new assay to determine single

nucleotide modification in DNA. In another assay wherein dark DNA-Ag clusters can

light up to bright red fluorescent clusters when placed in proximity of guanine rich

sequence of DNA.

Detection of Proteins

Metal nanoclusters have been proven to be excellent fluorescent protein sensors.

PAMAM encapsulated Nanocluster has been established as a detector of various protein

structures like, polyclonal goat driven antihuman antibody IgG, concanavalin A,

immunoglobulin G platelet drived growth factor AA, glutathione S-transferase tagged

protein etc.

Detection of other Biomolecules

Some other bio molecules with thiol functional group such as cysteine, homocysteine and

glutathione are very important biological analytes. In a certain bioassay Ag nanoclusters

are used for determination of cysteine among other amino acids. Fluorescence of Ag

nanoclusters can be successfully quenched with cysteine and not by other amino acids.

BSA-stabilised gold nanoclusters have been reported to detect glucose. The mechanism

of glucose oxidation involves production of H2O2 that can quench fluorescence of the

BSA-Au nanoclusters.

Part II

63

2.1.10 Chemosensing of Cu(I)

Since copper is one of the most abundant transition metal and is used in biological and

industrial systems. Selective recognition of copper has attained tremendous attention due

to its key role in several fundamental physiological processes including enzyme function

and transcriptional events. However elevated concentrations of copper ions in biological

systems can cause adverse health issues such as cytotoxicity and diseases like kidney and

liver damage and some neurodegenerative diseases are also caused by copper ions

imbalance (Scheinberg and Sternlieb 1996; Uauy, Olivares et al. 1998). Copper (I) ions

usually disproportionate rapidly in aqueous media and the most common example of

Cu(I) compound is cuprite ore (Cu2O) from which metallic copper is extracted. Due to

relative instability and low solubility of Cu(I) as compared to Cu(II), most of the

detection systems of copper are of Cu(II), which includes electrochemical (Yantasee,

Hongsirikarn et al. 2008), mass spectrometric (Siripinyanond, Worapanyanond et al.

2005), atomic absorption spectroscopic methods (Xiang, Zhang et al. 2010). Some

colorimetric assays are also recently reported (Kirubaharan, Kalpana et al. 2012; Deng,

Li et al. 2014) along with other optical systems as well.

2.1.11 Chemosensing of Fe(III)

Fe(III) is one of the most abundant metals on planet Earth and plays a significant role in

many biological and environmental systems, while it is the most abundant, vital transition

metal ion in the human body. As an essential part of heme groups, several proteins use

Fe(III) for oxygen and electron transportation, as well as for a variety of enzymatic

reactions (Thomson, Rogers et al. 1999). Fe(III) distribution in the human body is needed

to be critically regulated, due to its high potential for biological toxicity and many other

reasons. The cellular toxicity of Fe(III) ions has been associated with severe diseases,

including Alzheimer’s, and Parkinson’s disease (Andrews 2000). A lack of Fe(III) can

lead to anaemia, a disorder in which there are too few red blood cells, moreover, Fe(III)

deficiency limits oxygen delivery to cells, which results in fatigue, and decreased

immunity (Umbreit 2005). Conversely, excess of Fe(III) ions level in cells can catalyse

the production of reactive oxygen species (ROS), which can ultimately damage proteins,

Part II

64

nucleic acids and lipids (Tripathy, Woo et al. 2013). Furthermore, Fe(III) is a common

contaminant often found in the waste water released from the industrial sites. Fe(III) is

usually found in its more stable oxidized form that is Fe(III), which is nondegradable,

however the low solubility of Fe(III) does not give the necessary sensitivity for detection

in water.

Drinking water is a common source of heavy metal intake for humans. The toxicological

and carcinogenic effects of ultra-trace heavy metals can upset the central nervous system,

kidney, liver, skin, lungs, and bones (Kaplan, Yildirim et al. 2011). The detection and

removal of trace amounts of toxic metals in drinking water is an important factor in

monitoring environmental pollution and their adverse outcomes (El-Safty and Shenashen

2013).

Based on above discussion, the monitoring of metal ions traces in waters, soil surfaces,

and living beings, has become more and more important in recent times. Thus a variety of

methods have been developed for quantification of Fe(III) concentrations (Wang, Hai et

al. 2010; Wang, Yu et al. 2012; Mehta, Kailasa et al. 2014), flame atomic absorption

spectroscopy (FAAS) (Ajlec and Štupar 1989), ICPMS (Huang and Lin 2001), and

spectrophotometric detection using organic dyes or quantum dots(Wu, Li et al. 2009; Ju

and Chen 2014). To date, most Fe(III) sensing protocols depend on the fluorescence

quenching mechanism because of the paramagnetic character of ionic Fe(III), which

unavoidably give high background signals (Piao, Lv et al. 2014). The general drawbacks

of these techniques are that they often difficult to synthesize fluorescent probe, tedious

sample pretreatment, and sophisticated instruments (Annie Ho, Chang et al. 2012). The

selective optical sensing strategy has been striking due to its ultra-sensitivity, rapid

response, non-destructive nature and cost-effective manner (Bindhu and Umadevi).

2.1.12 Chemosensing of Pd(II)

Palladium contamination comes in environment mainly from two sources; one is by the

use of palladium in the catalytic converters of automobiles that release palladium, and the

other is by the residual palladium of synthetic intermediates in active pharmaceutical

products (Garrett and Prasad 2004; Magano and Dunetz 2011; Prichard and Fisher 2012).

Part II

65

Literature reveals that traces of palladium species in body organs interrupt a variety of

cellular processes and cause severe physiological disorders which includes loss of

memory, dizziness, migraine, immune system impairment, and paralysis (Kielhorn,

Melber et al. 2002). Therefore, the threshold level of palladium in pharmaceutical

products is strictly limited to be 5–10 ppm, while the maximum daily human dietary

intake of palladium is restricted to 1.5–15 mg (Carey, Laffan et al. 2006). It plays an

essential part in the manufacture of dental and medicinal devices, jewelry and automobile

(Iwasawa, Tokunaga et al. 2004). Palladium is known to bind with thiol containing

proteins, DNA and vitamin, then interferes the normal cell metabolism (Kielhorn, Melber

et al. 2002).

Palladium is a heavy transition metal that plays a critical role in synthetic materials and

chemical reactions. Palladium catalyzed reactions such as the Heck, Sonogashira, and

Suzuki- Miyaura reactions are turning increasingly important because of the influence of

making difficult covalent bonds. However, even after purification, residual palladium is

often found in the final product, which may cause health hazard (Song, Garner et al.

2007). Palladium compounds which are excessively used in the organic synthesis

reactions are Pd (PPh3)4, PdCl2, and Pd (OAC)2, while among these, PdCl2 is the most

toxic (Liu, Lee et al. 1979). Because of its toxicity but importance, the monitoring of Pd

traces in water, soil, plants, and physiologically important samples has become

increasingly important. Thus, the more traditional methods including AAS, ICP AES,

ICPMS, polarography, and voltammetry has been utilized for Pd(II) analysis over the past

few years (Wang and Varughese 1987; Zhao and Gao 1988; Locatelli 2006; Locatelli

2007; Gao, Koshizaki et al. 2009). But since all of these techniques in general require

expensive and dedicated equipments, tedious sample pretreatment, the use of toxic

reagents, and highly analytical expertise. Therefore, producing a facile, robust,

inexpensive, and sensitive method that permits real-time detection of Pd(II) ions is still a

challenging goal.

In recent years, several colorimetric and fluorimetric methods for detection of palladium

are reported because they are relatively more simple and nondestructive in nature

(Huang, Wang et al. 2004; Tang, Zhang et al. 2004; Schwarze, Müller et al. 2007; Song,

Part II

66

Garner et al. 2007; Duan, Xu et al. 2008; Jiang, Jiang et al. 2011; Zhou, Zhang et al.

2012). But most of the fluorescent sensors for palladium are based on the coordination

mechanism which are partially quenched due to the paramagnetic nature of the palladium

ion, and would suffer from varying degrees of interference from other transitional metal

ions. Among them, the reaction-based fluorescent sensing approaches have shown great

advantages in selectivity and sensitivity. However, these sensing systems have been

suffering from the demerits like synthesis of multi steps and expensive fluorescence

scaffolds, long response time, laborious test conditions etc., which greatly restrict their

practical applications. Therefore, the designing of some novel and effective chemosensor

for palladium detection with rapid response and high selectivity and sensitivity under

mild conditions is required for further developments (Ren, Pradhan et al. 2012).

2.1.13 Chemosensing of Antibiotics

6-Aminopenicillanic Acid (6-APA)

6-APA is the core of Beta lactam antibiotic class of compound penicillins. The penicillins

are being used commonly for their antimicrobial activity against both gram positive and

gram negative organisms (Wishart 1984). However, pencillins have limited stability, and

are susceptible to form different degradation products (Tyczkowska, Voyksner et al.

1992). The presence of degradation and related substances may cause the reduced activity

of the drug, as well as some side effects in humans. These antibacterial agents can

inactivate penicillin-binding proteins (PBP), which are used in the synthesis of bacterial

cell walls through the formation of stable covalent acyl complexes. As an outcome of this

irreversible substrate binding, the cell-wall-synthesizing activity of PBP is strongly

inhibited, which results into cell death. (Matagne, Lamotte-Brasseur et al. 1998; Fisher,

Meroueh et al. 2005). On the other hand, the occurrence of these antibiotics applied for

human or veterinary use in lakes and streams all over the world has created an alarming

situation due to the toxicity of these drugs for aquatic creature (Wilson, Smith et al. 2003;

Sanderson, Johnson et al. 2004). The common sources of their entry into the environment

include the excretion in urine, the wash off of topical treatments of livestock animals and

the pharmaceutical industrial unsafe waste. Hence, during our recent work of chemical

analyses of the drinking water system of Karachi, Pakistan we found a large number of

Part II

67

pharmaceuticals drugs in considerably high concentrations (Scheurell, Franke et al. 2009;

Selke, Scheurell et al. 2010).

Currently the most common analytical procedures which are employed for the detection

and/or determination of beta lactam antibiotics involve spectrometry (Abdalla 1991;

Devani, Patel et al. 1992), voltammetry (Ali and Mohammad 1994; Leszczynska, Koncki

et al. 1996), high-performance liquid-chromatographic (HPLC)(Pinto, Pavon et al. 1995),

(LC-MS) ( a -Cruz, López de Alda et al. 2003; Poole 2003), chemiluminescence

method(Yang, Zhou et al. 1998), and NMR (Shamsipur, Talebpour et al. 2002). Some

fluorimetric determination of beta lactamic antibiotics has also been described mainly

based on the formation of fluorescent degradation products either after alkaline or acid

hydrolysis (Murillo, Lemus et al. 1994; Yang, Zhou et al. 1998).

Pefloxacin (Pef)

Pef, 1-ethyl-6-fluoro-1, 4-dihydro- 7-(4-methylpiperazin-1-yl)-4-oxoquinolone-3-carboxylic

acid, is a fluoroquinolone antibacterial agent. Fluoroquinolones are effective against most

Gram-negative and -positive aerobic bacteria (Swoboda, Oberdorfer et al. 2003; Cao, Sui

et al. 2011), and also they have some activity against mycobacteria, mycoplasmas,

rickettsias and the protozoan Plasmodium falciparum (Sharma, Khosla et al. 1994). It is

used for the treatment for diseases of the skin and various kinds of urinary tract infections

and has been widely applied in clinical medicine (Ooishi and Miyao 1997).

Determination methods for Pef include spectrofluorometry (Veiopoulou, Ioannou et al.

1997; El-Kommos, Saleh et al. 2003), HPLC (Montay, Blain et al. 1983; Santoro, Kassab

et al. 2006), and capillary electrophoresis (Yang, Wang et al. 2008; Deng, Li et al. 2009),

nuclear magnetic resonanc e(Fardella, Barbetti et al. 1995), capillary electrophoresis

tandem mass spectrometry (Lara, García-Campaña et al. 2006), UV-vis (Mostafa, El-

Sadek et al. 2002; Sun, Wu et al. 2013), FT-IR(Xie, Song et al. 2010). However, these

methods require extensive sample preparation as well as highly trained individuals to

operate complicated instruments and understand complex chromatogram or spectral

results. Therefore, these traditional methods, although extremely accurate, are time-

consuming, expensive, and usually not suitable for use in the field. However, these

Part II

68

methods are often time-consuming or require substantial expertise in comparison with

spectrophotometry.

PEF had been used increasingly more in the aquaculture industry to treat and prevent a

variety of infectious diseases, but its overuse runs the risk of increasing drug residues in

seafood and contributing to bacterial resistance (Lu, Zhang et al. 2006). The common

sources of their entry into the environment include the excretion in urine, the wash off of

topical treatments of livestock animals and the pharmaceutical industrial unsafe waste.

Part II

69

2.2 RESULTS AND DISCUSSION

2.2.1Synthesis of Gold Clusters with Flexible and Rigid Diphosphine Ligands and

the Effect of Spacer and Solvent on the Size Selectivity

To study the effect of spacer on size selectivity, we selected few extremely wide range

geometrically ligands: a long chain, flexible alkyl diphosphine PPh2 (CH2)MPPh2 with M

= 6 i.e., 1,6-bis (diphenylphosphino)hexane denoted by L6, a completely rigid electronic

cloud rich xantphos denoted by LAr, another rigid ligand acetylenediphosphines (LAC),

and binap (LBn), and a moderately flexible spacer biphenyl (LBp) (Fig. 2.3).

Figure: 2.3 (A) 1,6-bis(diphenylphosphino)hexane denoted by L6, (B) Xantphos denoted by LAr, (C) Binap. LBn, (D) Biphenyl denoted by LBp, (e) Diphenylphosphinoacetylene

denoted by LAc

Furthermore we examined the effect of solvent on the core sizes of gold nanoclusters and

on the stability of molecular ion complexes by altering the solution from non-polar to

polar solvents.

(Polar) Methanol Ethanol Propanol ButanolCH2Cl2 CHCl3 CCl4 (Non-Polar)

2.2.1.1 Synthesis and Characterization of Bisphosphine Gold Nanoclusters

General procedure used for phosphine stabilized Au nanoclusters is similar to that

previously reported. Precisely, the mixture of one mole equivalent of Au (PPh3) Cl and

bisphosphine ligand is taken in a deaerated solvent of choice in an oven dried schlenk

flask under argon atmosphere. Vigorous stirring is continued and BTBC is added with

five mole equivalents as compared to gold precursor. The reaction mixture turned to

Part II

70

orange within 1-2 hours. Further stirring is continued for four hours to make sure the

completion of reaction. Products obtained were characterized by UV-Visible

spectrophotometry, and MALDI mass spectrometry. The main advantage of this one

phase, one pot synthesis system is its ease in handling and scaling up ability. To study the

effect of solvent polarities on stability and size selectivity of Au clusters, we synthesized

the Au clusters in methanol, ethanol, propanol, butanol, and the results are compared with

those synthesized in chloroform.

Characterization of L6 Au Clusters: The UV-Visible spectra recorded are shown in

Fig.2.4. It shows that in higher alcohols clusters of higher size are obtained and the

formed clusters are polydispersed. While in chloroform and methanol size selectivity was

better achieved. In the mixture of equal volumes of both chloroform and methanol two

bands were observed that describes the effects of solvent polarities on the size selectivity

of L6 Au clusters.

Part II

71

Figure 2.4 UV-Visible Spectrum of L6 Au Clusters in all solvents

MALDI spectrum of L6 Au Clusters revealed very interesting results (Fig.2.5). We

observed [Au8(L6)4]2+ at m/z 1695 while the intense peak at 1893 is designated to

[Au10(L6)4]2+. Other interesting peak appear at higher m/z ratio values at 2740 for clusters

of higher nuclearity like [Au21(L6)9]3+ corresponding to N>20 while another intense peak

is appeared at 3080 and is designated as cluster of highest nuclearity [Au40(L6)3]3+. We

also observed a chloro ligated cluster [Au16(L6)7Cl]2+ at m/z 3204.

Part II

72

Figure: 2.5 MALDI spectra of L6 obtained in all solvents

Characterization of LAr

Au Clusters:A comparative UV-Visible spectra is displayed in

Fig. 2.6. The best results here were obtained with chloroform. Absorption maxima at 418

nm show the formation of smaller clusters predominantly, and in alcohols we observed

clusters of higher nuclearity. Here again in the mixture of chloroform and methanol we

observed two prominent absoption bands which represents the size selectivity as a

function of solvent polarities.

Part II

73

Figure: 2.6 UV-Vis spectrum of LAr Au Clusters in all solvents used

MALDI analysis shown in Fig. 2.7 gives the clear comparison of solvents effect on

selectivity of clusters produced. The formation of [Au9(LAr)6]3+ is observed at 1747 and a

comparatively less intense peak corresponds to the formation of [Au10(LAr)4]2+ which

appeared at 2141. A less intense peak was observed, indicating the formation of high

nuclearity complex [Au29(LAr)3]3+ at m/z 2482 in ethanol and butanol. In most polar

solvent of the study methanol, greater selectivity is observed for smaller clusters. We

observed a peak at m/z 1448 in all the solvents and is assigned to any gold cluster

[AunLArn]n+.

Part II

74

An interesting approach was used to conclude the effect of polarities of solvents on

size selectivity of nanoclusters by taking the MALDI spectrum in 1:1 mixture of

chloroform and methanol. This result led us to a tremendous technique that can be

used to optimize the best suitable solvent to get the core size of desired nuclearity, the

only clusters obtained was Au9 and Au10. When the reaction was carried out in

butanol, [AunLArn]n+ was observed as most abundant peak, however partial ligand

elimination gave an almost equal abundant peak at m/z 1408 and is assigned to

[Au3LArP((C6H3)2OC(CH3)2)]+.

[AuxLAry]z+

[Au3LArP((C6H3)2OC(CH3)2)]+ + PPh3 + [Au2LArPh]+

To further investigate the effect of polar solvents we investigated higher homologues

of alcohols, the results led us to the formation of [AunLArn]n+, and relatively small

peak of Au9 is also obtained, while partial ligand dissociation gave two fragments of

low intensity at m/z 1090 and 1408 and are corresponded as

[AuxLAry]z+

[Au3P((C6H3)2OC(CH3)2)PPh3]+ + [Au3LArP ((C6H3)2OC(CH3)2)]++PPh3

Part II

75

Figure 2.7 MALDI Spectral Ananlysis of LAr Ligated Au Clusters in different solvents

Characterization of LAc

Au Clusters: In the case of most smallest and rigid ligand of the

study we observed that pronounced selectivity is achieved in alcohols while absorption at

higher wavelength in chloroform suggest that non polar medium does not favour the

formation of small cluster. UV-Vis spectrum is given below (Fig. 2.8)

Part II

76

Figure: 2.8 UV-Vis Spectra of LAc Au Clusters in all Solvents

MALDI analysis (Fig. 2.9) shows that clusters of variable size are obtained in this case.

We observed the peak of [Au9L3]3+ at m/z 985 in all the solvents, while signal at m/z 1643

is assigned to [Au13L6]3+. [Au14L6Cl]2+ is obtained in chloroform and in methanol and is

observed at m/z 2560. While Au19 as [Au19L2Cl2]2+ was also observed at m/z 2990 in only

propanol.

Part II

77

Figure: 2.9 MALDI Spectral Ananlysis of LAc Ligated Au Clusters in different solvents

Characterization of LBn

Au Clusters: The most selective solvents for LBn for smaller

clusters for example Au9 and Au10 were found to be methanol and mixture of chloroform

and methanol as compared to other alcohols since they showed a broad absorption band

in the range of higher wavelengths as depicted in Fig. 2.10 below.

Part II

78

Figure: 2.10 UV-Vis Spectra of LBn Au Clusters in all solvents

MALDI spectra of LBn ligated Au Clusters is shown in Fig. 2.11. In methanol it gave the

formation of [Au9L3]3+ at m/z 1213 along with the [Au9L6]3+ at 1835. We also found

[Au10L4]2+ and [Au14L4]2+ as moderately intense peaks at m/z 2229 and 2623 respectively.

The MALDI spectrum of clusters synthesized in chloroform gave largely the Au10, and

when these two results are compared with the one synthesized in mixture of both solvents

we obtained appreciable peaks of Au9, Au10 along with less intense Au13 and Au14 those

were also found in chloroform but not in methanol. The intensity of higher nuclearity

cluster that is [Au14L4]2+ and [Au14L6]2+ were greater in chloroform as compare to

methanol, and were not formed in other alcohols. In propanol we detected [Au13L6]3+ as a

moderate abundant signal while ethanol showed selectivity for Au13 and butanol for Au14.

Part II

79

Figure: 2.11 MALDI Analysis of LBn Au Clusters in all Solvents

Part II

80

Characterization of LBp

Au Clusters: UV-Vis spectra of LBp Ligated Au clusters are

displayed in Fig. 2.12.

It is evident that smaller clusters are selectively formed in chloroform, methanol, ethanol

and the mixture of chloroform and methanol because their absorption maxima lie within

the range of 410 to 450 nm. The best results are obtained with chloroform, it is showing a

narrower band at about 420 nm which suggest the formation of monodisperse small Au

clusters. In other solvents we also observed that another broader absorption band arise in

the higher wavelengths range which describes those chances of polydispersity of larger

clusters in this case.

Part II

81

MALDI spectra of LBp Au clusters (Fig. 2.13) explains that, smaller cluster [Au8L5]2+ is

obtained in chloroform and methanol at m/z 2094 as suggested by UV-Vis spectra, while

we also observed [Au8L5Cl]3+which appeared at m/z 1407 in all solvents except propanol

and butanol, and its absorption intensity is greatest in chloroform and when the spectra is

recorded in mixture of chloroform and methanol it gave a small peak. Au9 is formed in all

solvents and is observed at m/z 1635 as [Au9L6]3+, and the most abundant signal was

observed in mixture of chloroform and methanol as compared to others. [Au10L4]3+ is also

formed in low amounts and is obtained at m/z 2029. A higher nuclearity cluster is formed

in propanol, butanol and in mixture of chloroform and methanol that is [Au14L6]2+ which

appeared at m/z 2945. Although methanol and ethanol are found to be very selective for

the formation of Au9 and Au10.

Figure: 2.13 MALDI Analysis of LBp Au Clusters in all Solvents

Part II

82

2.2.2 Atomic Force Microscopic guided detection of Copper in real samples and T-

lymphocytes with Pyridinium Thioacetate capped Silver Nanoparticles

Pyridinium compounds are known to form ionic liquids (ILs) which have been

extensively used as stabilizing/capping agents for metal nanoparticles (Dupont, Fonseca

et al. 2002; Kim, Demberelnyamba et al. 2003). Reports show that these systems usually

tend to cause aggregation of nanoparticles due to instability and their phase transfer

characteristics (Cassol, Umpierre et al. 2005; Wang and Yang 2005). The uses of

pyridinium ligands are not only limited to ILs but their wide spread occurrence in nature

make them fascinating candidate for biological and photophysical evaluation. Several

pyridinium nanoconjugates and complexes have recently being developed for catalysis

(Tourneux, Gornitzka et al. 2007; Zhu, Jang et al. 2011), drug delivery, biocidal activities

(Patil, Kokate et al. 2011) and molecular recognitions (Saha, Agasti et al. 2012;

Bagherzadeh, Pirmoradian et al. 2014).

2.2.2.1 Synthesis of 4-formylpyridiniumpropylthioacetate Coated Silver

Nanoparticles (4-Py-AgNPs)

1 mL of 0.1 mM solution of 4-Py was added in a shaking solution of 10 mL of 0.1 mM

silver nitrate. After 15 minutes, 0.1 mL of freshly prepared 4 mM NaBH4 solution was

added in the reaction mixture. The colorless reaction mixture turned yellow-brown

instantly after addition of NaBH4 solution, but the solution was stirred further for 2 hours.

The AgNPs colloidal dispersions were centrifuged for 20 min till all nanoparticles

precipitated at 10,000 rpm to separate the potential large aggregates and excess unbound

ligand from the broth followed by redispersion in deionized water. The yellow

supernatant was freeze dried and further used whenever required. The resulting solution

was subjected to UV-visible spectrophotometry and AFM imaging.

2.2.2.2 Characterization of 4-Py-AgNPs

Upon contacting the 0.1 mM solution of 4-Py with 0.1 mM AgNO3 and 4 mM NaBH4

solution, the UV–vis absorption spectra of the Ag nanoparticle showed surface plasmon

band (SPR) at 430 nm (Fig. 2.14) indicating that the reaction is spontaneous. The SPR

Part II

83

band steadily increased in intensity and finally reached to maximum after lapse of 30

minutes. However, the color change of the reaction mixture from colorless to dark brown

was immediately observed. The color change is mainly due to the excitation of surface

plasmon vibrations in Ag nanoparticles.

300 400 500 600 7000.0

0.1

0.2

Ab

sorb

ance

(un

its)

Wavelength(nm)

0.05mM AgNO3

AgNP

0.05mM Ligand

Figure: 2.14 UV-visible spectra of the AgNPs

Thermal stability of the colloidal solution was determined by boiling the nanoparticles at

100 ºC for 2 hours. Fig. 2.15 indicate that there is no change in the absorption band after

heating.

Part II

84

300 400 500 600 7000.0

0.1

0.2

Ab

sorb

an

ce(u

nit

s)

Wavelength(nm)

Boiled

Unboil

Figure: 2.15 UV-visible spectra of the AgNPs solution at room temperature and

boiling point

The effect of electrolytes on the stability of colloidal silver was investigated by mixing

different concentrations of aqueous NaCl solutions (10 uM, 50 uM, 1mM, 5mM, 10 mM,

50 mM, 100 mM, 500 mM and 1 M) with 0.05 mM AgNPs solution and the spectra are

shown in Fig. 2.16. The SPR band of colloidal AgNPs unaffected till mixing of 50 mM

concentration of NaCl but mixing of higher concentration of salt solution led to the

agglomeration of colloids.

Part II

85

300 400 500 6000.0

0.1

0.2

Ab

sorb

ance

(un

its)

Wavelength(nm)

ZAgNp(50uM)

Z+10uM NaCl

Z+50uM NaCl

Z+1mMNaCl

Z+5mMNaCl

Z+10mMNaCl

Z+50mM NaCl

Figure: 2.16 UV-visible spectra of salt addition stability of AgNPs

2.2.2.3 Chemosensing of Cu(I) by 4-Py-AgNPs

The 4-Py-AgNPs possesses a quaternary nitrogen and benzaldehyde. The aldehyde group

of the ligand can interact with metal cations so a number of metal cations (50 µM) such

as, Cu(I), Ca(II), Zn(II), La(III), Sb(III), Fe(III), Co(II), Pd(II), Rb(I), Y(III), Cu(II), and

Na(I)were titrated against AgNPs (50 µM) and the interaction was evaluated with the

help ofUV spectrophotometer (Evolution 300). The UV visible titration profile of

AgNPsbefore andafter mixing of selected metal cations (Fig.2.17A) revealed selective

interaction of AgNPs with Cu(I). A decline in the absorption intensity of AgNPs was

observed at 403 nm after contacting it with Cu(I). Reduction in theabsorbance of

AgNPsis attributed to its selective binding and accessibility for Cu(I) ions via an

intermolecular complexation. The Cu(I)is believed to be engulfed by the chelating

functional groups of the AgNPs.

Part II

86

280 350 420 490 560 6300.00

0.04

0.08

0.12

0.16

0.20

ZAgNPs

ZAgNP+Cu(I)

ZAgNP+Ca(II)

ZAgNP+Co(II)

ZAgNP+Fe(III)

ZAgNP+La(III)

ZAgNP+Na(I)

ZAgNP+Ni(II)

ZAgNP+Pd(II)

ZAgNP+Rb(I)

ZAgNP+Y(III)

ZAgNP+Zn(II)

ZAgNP+Cu(II)

ZAgNP+Sb(III)Ab

sorb

ance

(un

its)

Wavelength(nm)

A

4030.0

0.1

0.2

Ab

sorb

ance

(un

its)

Wavelength(nm)

ZAgNPs

ZAgNPs+Cu(I)

ZAgNPs+Ca(II)

ZAgNPs+Co(II)

ZAgNPs+Fe(III)

ZAgNPs+La(III)

ZAgNPs+Na(I)

ZAgNPs+Ni(II)

ZAgNPs+Pd(II)

ZAgNPs+Rb(I)

ZAgNPs+Y(III)

ZAgNPs+Zn(II)

ZAgNPs+Cu(II)

ZAgNPs+Sb(III)

B

Figure: 2.17 A) UV-visible spectra of metal cations after titrations with nanoparticles. B)

Bar graph representation of metal cations after titrations with nanoparticles at λmax

Part II

87

Functional groups of 4-Py-AgNPs were deduced after evaluation of FTIR spectrum of

AgNPs (Fig. 2.18). The stretching band at 1350-1400 cm-1 confirms the presence of

sulfur. Stretching at 668cm-1 was attributed to C-S bond. As soon as the AgNPs are

treated with Cu(I) a bathochromic shift in the stretching of C-S was observed which also

indicated the tendency of Cu(I) to form metallic bond with Ag.

Figure: 2.18: A) FT-IR spectrum of 4-Py AgNPs

Part II

88

Figure: 2.18 B) FT-IR spectrum of 4-Py AgNPs after treating with Cu(I)

The size and morphology of the nanoparticles were established with atomic force

microscope (AFM). As shown in Fig. 2.19 that the nanoparticles are spherical in shape

and its size is in the range of 5-10 nm. It is evident from Fig. 2.19 that the nanoparticle

size decreases after addition copper`s solution. Reduction in size of AgNPs after mixing

with Cu(I) solution could be attributed to change in aqueous electrical conductivity of the

colloids due to addition of salt solution which leads the particles to further split into

smaller size to overcome the equilibrium disturbance (Jang, Ortega et al. 2012). Another

possible explanation for the size reduction of AgNPs after interactions with Cu(I)

solution may be alloying of metal ions which can link the two metals and hence contracts

the particle size (Valodkar, Modi et al. 2011).

Part II

89

Figure: 2.19 a) Topographical AFM image of 4-Py AgNPs b) Topographical AFM

image of AgNPs after mixing with Cu(I)

Part II

90

The kinetics of interactions between AgNPs and Cu(I) solutions were studied by

recording absorption spectra of the soup bearing AgNPs and Cu(I) at various interval of

time As shown in Fig. 2.20 that immediate change in the SPR band of AgNPs was

observed.

Figure 2.20 Kinetic studies after interaction of Cu(I) with4-Py AgNPs

The synthesized AgNPs was found to be stable in basic media while in acidic media (pH

1-7) the colloids were not stable and prone to aggregation. The results are summarized in

Fig. 2.21. Peak broadening is observed at pH lower than 7 which is in line with literature

reports (Mulvaney 1996). Prathna et al. described that increase in pH of citrate stabilized

AgNPs lead to blue shift due to reduction of particle size (Prathna, Chandrasekaran et al.

2011) at lower pH.

Part II

91

The effect of pH on the interaction of AgNPs with Cu(I) was evaluated. The Fig. 2.21

clearly shows that best interactions between AgNPs and Cu(I) is observed around neutral

pH. The acidic pH can protonate the electron donating groups of the ligand coated around

AgNPs thus decreasing its interaction with Cu(I). Similarly under highly basic conditions

the Cu(I) can precipitate out hence decreasing the concentration of Cu(I) in the media.

Figure: 2.21: Effect of pH on the selective recognition of Cu(I) by AgNPs.

Sensitivity is an important factor that should be evaluated while studying an acceptable

chemosensor. A 50 μM aqueous solution 4-Py AgNPs was titrated against various

concentrations of Cu(I) at pH 8. Absorption spectra (Fig. 2.22) revealed that mixing of

Cu(I) with a 50 μM aqueous solution of 4-Py AgNPs caused a gradual and smooth

decrease in the absorption intensity of 4-Py AgNPs upon gradual increase in the

concentration of Cu(I).

Part II

92

The limit of detection of 4-Py AgNPs as a chemosensor for the detection of Cu(I) was

deduced from the plot of absorption intensity as a function of the concentration of Cu(I).

The lowest limit of detection found to be 1 µM with a regression coefficient of 0.99.

Figure: 2.22 A) Selective recognition of Cu(I) by 4-Py AgNPs at different concentrations B)

Linear regression curve obtained after treating varies concentration of Cu(I) with 4-Py AgNPs.

Part II

93

The affinity of AgNPs toward Cu1+ was studied in the presence of competing metal

cations discussed earlier. The absorption intensity of AgNPs was monitored after titrating

mixture of AgNPs (50 µM) and Cu1+ (50 µM) against 50 µM aqueous solution of each

metal cations such Ca2+, Cu2+, Co2+, Fe2+, Na1+, Ni2+, Sb3+, Y3+, Zn2+, Rb1+ and Pd2+.As

shown in Fig. 2.23 that thecompeting metal cations didn`t interfere in the absorption

behaviour of AgNPs-Cu1+complex. It is established that AgNPsis discriminating Cu1+ in

the presence of other competing metal cations.

280 350 420 490 560 6300.00

0.04

0.08

0.12

0.16

0.20

Abso

rban

ce(u

nits

)

Wavelength(nm)

ZAgNP

ZAgNP+Cu(I)+Water

ZAgNP+Cu(I)+Ca(II)

ZAgNP+Cu(I)+Co(II)

ZAgNP+Cu(I)+Cu(II)

ZAgNP+Cu(I)+Fe(III)

ZAgNP+Cu(I)+La(III)

ZAgNP+Cu(I)+Na(I)

ZAgNP+Cu(I)+Ni(II)

ZAgNP+Cu(I)+Sb(III)

ZAgNP+Cu(I)+Y(III)

ZAgNP+Cu(I)+Zn(II)

ZAgNP+Cu(I)+Rb(I)

ZAgNP+Cu(I)+Pd(II)

A

4040.00

0.02

0.04

0.06

0.08

0.10

0.12 ZAgNP

ZAgNP+Cu(I)+Water

ZAgNP+Cu(I)+Ca(II)

ZAgNP+Cu(I)+Co(II)

ZAgNP+Cu(I)+Cu(II)

ZAgNP+Cu(I)+Fe(III)

ZAgNP+Cu(I)+La(III)

ZAgNP+Cu(I)+Na(I)

ZAgNP+Cu(I)+Ni(II)

ZAgNP+Cu(I)+Sb(III)

ZAgNP+Cu(I)+Y(III)

ZAgNP+Cu(I)+Zn(II)

ZAgNP+Cu(I)+Rb(I)

ZAgNP+Cu(I)+Pd(II)

Abso

rban

ce(u

nits

)

Wavelength(nm)

B

Figure: 2.23 A) Absorption spectra of 4-Py AgNPs+Cu(I) in presence of other

competing metal cations B) bar graph representation Absorption spectra of 4-Py AgNPs+Cu(I) in presence of other competing metal cations

Part II

94

Stochiometry of interaction of AgNPs with Cu(I) was carried out through Job’s plot

experiment. It is shown in Fig. 2.24 that absorbance maxima intersect at the 0.5 mole

fraction on the x-axis (mole fraction). It implies that the complexation ratio between

AgNPs and Cu(I) is 1:1.

Figure: 2.24 Complexation ratio between 4-Py AgNPs and Cu(I) The analytical potential of the AgNPswas evaluated by using it for selective detection and

quantification of Cu1+ in tape water of Karachi University. Tape water samples from

University of Karachi were spiked with50 µM Cu1+. The AgNPs proved to be valuable probe

for detection of Cu1+ with lower limit of detection 2 μM, which is much lower than the

maximum allowed limit (∼20 μM) of Cu1+ in drinking water permitted by the U.S.

Environmental Protection Agency (EPA) (G. Georgopoulos 2001). Nearly similar results

were obtained when tape water was used instead of deionized water for preparation of

copper`s solution and titrated against AgNPs (Fig. 2.25). UV-visible guided titrations of

AgNPs (50µM) with Cu1+ (50 µM) spiked in human blood plasma lead to reduction of

absorption intensity (400 nm) with the increase of Cu1+ concentration (Fig. 2.25). The

obtained decline in absorption intensity is directly proportional to the increase of Cu1+

concentration in a wide range (i.e. 2-50 µM), that is inline with the copperconcentration

range in healthy body plasma samples (Malavolta, Giacconi et al. 2010; Fayet-Moore, Petocz

et al. 2014). This shows that AgNPs can be a valuable tool which shows the plasma sample is

healthy or not.

Part II

95

300 400 500 600 700

0.0

0.1

0.2

0.3

Abs

orba

nce(

unit

s)

Wavelength(nm)

Tap water

CuCl in tap water

AgNPs

AgNPs + CuCl in tap water

A

300 400 500 600 7000.0

0.5

1.0

1.5

Abs

orba

nce(

unit

s)

Wavelength(nm)

AgNPs

Plasma

AgNPs+Cu+Plasma

B

Figure 2.25 UV-visible spectra of 4-Py AgNPs recorded in presence Cu(I) spiked in (A)

Human plasma (B) Tap water

Part II

96

2.2.2.4 Intracellular Cu(I) detection in T-lymphocytes:

Cell incubation

Cytotoxic T lymphocytes (CTLL-2) from (ECACC, ATCC) for AFM based study were

used. For this purpose, the CTLL-2 (ATCC® TIB-214™) cells was cultured in ATCC-

formulated complete RPMI 1640 ((1mM Sodium pyruvate, 2M HEPES and 10% FBS,

10% T-STIM & Con A, 1, 1% L-glutamine and Non-Essential Amino Acids) culturing

media in 75 cm2 flasks, and kept in 5% CO2 incubator at 37 oC. After complete

confluences about 5×104 cell/well (in 96 well flat bottom plate corning) of cell line were

used to conduct the experiments.

Cell Treatment

The cell permeability of the AgNPs on T-cells was evaluated by incubating AgNPs at

different doses with T-cells for 30 min at 37 oC in 5% CO2 incubator. After incubation

the cells were centrifuged at 1300 rpm for 4 min and the cell pellet were washed twice

with 1X, PBS (Phosphate buffer saline) buffer to remove the unbound test sample. Then

the treated cells were incubated for 20 min with multiple concentration of Cu+ solution at

37 oC in 5% CO2 incubator followed by AFM microscopy.

MTT Based Cytotoxicity Assay

Cytotoxicity of test samples were evaluated in 96-well flat-bottomed micro plates by

using the standard MTT (3-[4,5-dimethylthiazole-2-yl]-2, 5-diphenyl-tetrazolium

bromide) colorimetric assay. For this purpose, the lymphocytes cells were cultured in

their respective media RPMI 1640 in 75 cm2 flasks, and kept in 5% CO2 incubator at 37 oC. Cell culture with the concentration of 5x104cells/ml was prepared and introduced

(100 µL/well) into 96-well plates. After overnight incubation, medium was removed and

200 µL of fresh medium was added with different concentrations of test sample (30,

50,100 µM). After 48 hrs, 200 µL MTT (0.5 mg/ml) was added to each well and

incubated further for 4 hrs. Subsequently, 100µL of DMSO was added to each well. The

extent of MTT reduction to formazan within cells was calculated by measuring the

absorbance at 540 nm, using a micro plate reader (Spectra Max plus, Molecular Devices,

CA, USA). The percent inhibition was calculated by using the following formula:

Part II

97

Inhibition (%) = [1 - (treated/control)] × 100

Viability (%) = 100 – inhibition (%)

The potential of nanoparticles to selectively detect metal cations in living cells is vitally

important for biological application. Taking into that copper accumulation in the body

leads to toxicity, we primarily conducted investigation using AgNPs on T-cell lines in

which copper is known to be accumulated To determine the cell permeability of AgNPs,

T cells were supplemented with multiple doses of AgNPs in the range of 10-0.5 μM for

10-20 min at 37 °C, and washed with PBS to remove the surplus AgNPs. One can clearly

observe significant accumulation of nanoparticles inside the cells, and the cells are still

intact, indicating permeability of nanoparticles through cell membrane. At 1 μM lower

concentration of AgNPs, the nanoparticles are appeared as small grooves on the cell

surface but at higher concentration, the cells are fully loaded with nanoparticles and they

are still in good health and shape as in the case of 10 μM. While images were taken upon

the addition of CuCl (5, 10, 30, 40 and 50 μM, respectively) at 37 °C for 20 min. In Fig.

2.26, an untreated T-cell image is shown which is used as control throughout the

microscopic studies. We have distributed the host guest intracellular interactions in three

categories. First at lower concentrations of both the guest and host (for example at 0.5

μM nanoparticles and 5 μM), the morphology of cell surface is shrinked by the

interaction between the two. While even on addition of more copper (10 μM) the cells are

not broken (Fig. 2.26). Similar results were obtained when we increased the nanoparticles

and copper concentration to 1 μM and 30 μM respectively (Fig. 2.27). Then at higher

doses (10 μM nanoparticles, we observed a few interesting things. First, the nanoparticles

are completely inside the cell, and they quenched the cytotoxicity at lower concentration

of copper that is 5 μM. Secondly, at higher doses of copper (50 μM), its starts to outweigh

the effect of nanoparticle interaction and a disrupt in cell morphology can be seen in Fig.

2.28 along with a little or no effect on two different cells. This effect of concentration is

completely supported by MTT cytotoxicity assay results given below.

The MTT cytotoxicity assay results suggest that when cells were treated with varying

concentrations of copper, the cell viabilities are found to be 12, 20, and 26 % at 30, 40,

and 50 μM respectively. While AgNPs did not exert any adverse effect on cell viability,

and the viabilities are recovered to 53 % and 47 % at 10 and 1 μM concentrations of

Part II

98

AgNPs respectively. The effect was more pronounced in higher concentration and

showed the recovery of cells viability in a dose-dependent manner.

Figure: 2.26 A) T cell (Control)

Figure: 2.26 B) 0.5 μM 4-Py AgNPs treated T cell

Part II

99

Figure: 2.26 C) 0.5 μM 4-Py AgNPs treated T cell with 5 μMCu (I), cell

surface is shrinked due to interactions.

Part II

100

Figure: 2.26 D) 0.5 micromolar 4-Py AgNPs treated T cell with 10 micromolar Cu (I)

Part II

101

Figure: 2.27 A) 1 micromolar 4-Py AgNPs treated T cell with 5 micromolar Cu (I)

Figure: 2.27 B) 1 micromolar 4-Py AgNPs treated T cell with 30 micromolar Cu (I)

Part II

102

Figure: 2.28 A) 10 micromolar 4-Py AgNPs treated t cell

Figure: 2.28 B) 10 micromolar 4-Py AgNPs treated t cell with 5 micromolar copper

Part II

103

Figure: 2.28 C) 10 micromolar 4-Py AgNPs treated t cell with 50 micromolar copper

Part II

104

2.2.2.5 Chemosensing of 6-AminopenicillanicAcid by 4-Py-AgNPs

6-Aminopenicillanic acid (6-APA) is the core of Beta lactam antibiotic class of compound

penicillins. The penicillins are being used commonly for their antimicrobial activity against

both gram positive and gram negative organisms (Wishart 1984). However, pencillins have

limited stability, and are susceptible to form different degradation products(Tyczkowska,

Voyksner et al. 1992). The presence of degradation and related substances may cause the

reduced activity of the drug, as well as some side effects in humans. These antibacterial

agents can inactivate penicillin-binding proteins (PBP), which are used in the synthesis of

bacterial cell walls through the formation of stable covalent acyl complexes. As an

outcome of this irreversible substrate binding, the cell-wall-synthesizing activity of PBP is

strongly inhibited, which results into cell death.(Matagne, Lamotte-Brasseur et al. 1998;

Fisher, Meroueh et al. 2005). On the other hand, the occurrence of these antibiotics applied

for human or veterinary use in lakes and streams all over the world has created an alarming

situation due to the toxicity of these drugs for aquatic creature (Wilson, Smith et al. 2003;

Sanderson, Johnson et al. 2004). The common sources of their entry into the environment

include the excretion in urine, the wash off of topical treatments of livestock animals and

the pharmaceutical industrial unsafe waste. Hence, during our recent work of chemical

analyses of the drinking water system of Karachi, Pakistan we found a large number of

pharmaceuticals drugs in considerably high concentrations (Scheurell, Franke et al. 2009;

Selke, Scheurell et al. 2010).

Currently the most common analytical procedures which are employed for the detection

and/or determination of beta lactam antibiotics involve spectrometry(Abdalla 1991;

Devani, Patel et al. 1992), voltammetry (Ali and Mohammad 1994; Leszczynska, Koncki et

al. 1996), high-performance liquid-chromatographic (HPLC) (Pinto, Pavon et al. 1995),

(LC-MS) ( a -Cruz, López de Alda et al. 2003; Poole 2003), chemiluminescence method

(Yang, Zhou et al. 1998), and NMR (Shamsipur, Talebpour et al. 2002). Some fluorimetric

determination of beta lactamic antibiotics has also been described mainly based on the

formation of fluorescent degradation products either after alkaline or acid hydrolysis

(Murillo, Lemus et al. 1994; Yang, Zhou et al. 1998). However, all these reported methods

have their own disabilities and limitations, since the fluorescence probes are difficult to

Part II

105

design and involve a tedious protocol of purifying the molecules and comprise highly

skilled synthetic expertise.

Nanoparticles synthesis and their applications in chemosensing have grabbed lots of

interest especially in biosensing of metals and drugs (Jung, Lee et al. 2011; Saha, Agasti et

al. 2012; Liu, Zhou et al. 2014). Nanoparticles probes have tremendous advantages over

other methods, due to their simplicity, cost effectivity and robustness. Recently ateeq etal

reported a natural product capped AuNPs for the chemosensing of piroxicam (Ateeq, Shah

et al. 2014), and in another report yang etal published a tetracycline detection system based

on nanomaterial probe (Yang, Zhu et al. 2014). Inspired by these reports, we put our

attention towards developing a beta lactam antibiotic nanosensor.

Application of 4-PyAgNPs as 6-APA Sensing Probe

The recognition behavior of fully characterized 4-PyAgNPs was evaluated for various

drugs by the 1:1 mixture of 0.05 µM drugs solutions and AgNPs. The UV–vis spectra of

AgNPs were recorded instantly after addition of drug solutions. The effect of tested drugs

such as 6-APA, amoxicillin, aspirin, cefaclor, cefotaxime sodium, ceftriaxone sodium,

cephalexime, cephradine, diclofenac sodium, paracetamol, pefloxacin, and penicillin on

SPR band absorption intensity was investigated and plotted in Fig. 2.29. All drugs

decreased the absorbance intensity of 4-PyAgNPs, but the maximum quenching in SPR

band is observed with 6-APA. The slight decrease in absorbance of AgNPs is associated

with the dilution as in case of all the drugs, but the considerable large difference between

rest of the drugs and 6-APA absorption behavior indicates some chemical or physical

interactions. These results of spectrophotometric drugs screening can be exploited as a

selective assay towards 6-APA detection.

Part II

106

350 400 450 500 5500.00

0.05

0.10

0.15

0.20

Abs

orba

nce(

unit

s)

Wavelength(nm)

0.05mM ZAgNp

ZAgNp+6APA

ZAgNp+Amoxillin

ZAgNp+Aspirin

ZAgNp+Cefaclor

ZAgNp+CefotoximeSodium

ZAgNp+CeftrixonSodium

ZagNp+Cephalexime

ZAgNp+Cephradine

ZAgNp+Diclofinic

ZAgNp+Paracetmol

ZAgNp+Pefloxime

ZAgNp+Penicillin

Figure: 2.29 Drugs screening study

AFM and FT-IR spectral analysis to understand the interactions between 4-PyAgNPs

and 6-APA

The interaction of nanoparticles with other organic species like metal ions and drugs may

cause some morphological change. Some recent reports indicate the decrease in

nanoparticles size upon interaction with other chemical entities (Pingali, Deng et al.

2008; Ohkubo, Seino et al. 2014). We also observed the size of 4-PyAgNPs changed

largely from 40-60 to 0.1-5 nm (Fig. 2.30). This behavior in the size domain of

nanoparticles is explained by comparative FT-IR spectral studies.

The comparative FT-IR spectral analysis is carried out to have a deeper understanding of

the mechanism of nanoparticles formation and their 6-APA recognition. Ligand FT-IR

spectrum shows the C-S stretches at 621 and 1365 1/cm, while thioester carbonyl is

appeared at 1638. As it is very well established fact these days, we also observed that the

thioester peak is diminished upon the formation of AgNPs which is an indication that

Part II

107

stabilization of AgNPs is occurred directly by Sulfur. We also recorded the FT-IR spectra

of 6-APA, which showed all the characteristics peaks of the drug including lactam

carbonyl at 1623, acid carbonyl at 1773 and N-H stretchings at 2904 and 2987 1/cm,

while due to carboxylic hydroxyl group, the whole region around 2900-3300 1/cm

showed a broad signal. Interestingly, when 6-APA was treated with 4-Py-AgNPs, the

acid carbonyl peak which was appeared at 1773 1/cm in the drug was absent here. These

results describes that, in the slightly basic pH of the colloidal suspension the carboxylic

acid group shows hydrogen bonding with free aldehydes on 4-Py AgNPs surface. Due to

hydrogen bonding interactions, acid carbonyl peak is broadened and merged with that of

lactam carbonyl, as well as a broad signal of hydroxyl (H-bonding) is appeared at 3412

1/cm.

Figure: 2.30 Morphological change in the nanoparticles after interaction with 6-APA

Effect of the pH on the Stability of 4-Py AgNPs and 6-APA Recognition

Since the pH of the medium has a great influence on the optical response of the

complexation reaction, an optimum pH is thus essential to develop for the interaction of

4-Py AgNPs-6APA complex. A study of the pH in the range of 2–12 with the optimized

conditions for interaction of nanoparticles with the drug is shown in Fig. 2.31. The

Part II

108

absorbance of SPR band of the AgNPs-6APA mixture shows the maxima at pH 9, and

before and after which it gradually decreases. Moreover, it can be seen with the

comparative pH plots that the interaction of drug and nanoparticles follows the trend of

the buffer stability of the nanoparticles with subsequent quenching in the absorbance of

SPR band. Therefore on the basis of these results it can be safely concluded that the drug

and nanoparticles interaction is independent of the pH change, and our system works

almost equally well at most of the pHs.

Figure: 2.31 Effect of pH on the stability of 4-Py AgNPs and accumulation of

nanoparticles with 6-APA

Analytical Performance of the Nanosensor

The performance of our optimized detection system is demonstrated by evaluation of the

concentration studies. We used various concentrations of 6-APA to measure the sensitive

response of developed analytical system. Fig. 2.32 represents that the data strictly

followed beer’s law and exhibited a good linear correlation to 6-APA concentration in the

range from 1 to 50 μM with the regression constant (R2) equal to 0.992, while the limit of

detection (LOD) is found to be 19.08μM. Moreover, to estimate the binding

stoichiometry between AgNPs and 6-APA Job's plot experiment was performed.

0 0.02 0.04 0.06 0.08

0.1 0.12 0.14 0.16 0.18

0 1 2 3 4 5 6 7 8 9 10 11 12

Ab

sorb

ance

(un

its)

pH(units)

ZAgNp

APA

Part II

109

Absorption spectra were recorded by varying the mole ratio of 6-APA to AgNPs. The

minimum on the plot between absorbance and mole fraction is appeared at 0.5, which

correspond to the 1:1 stoichiometric relation between the stable AgNPs-6APA

complexes.

Part II

110

Figure: 2.32 A) UV-visible spectra of concentration study, B) Calibration

curve and C) Job’s plot

Specificity

The specificity of this nanosensor was tested by evaluating the response of optimized

system towards other interfering drugs. All the drugs were used in 1:1 (v/v) with 6-APA.

Fig. 2.33 shows that the additions of competing drugs such as amoxicillin, aspirin,

cefaclor, cefotaxime sodium, ceftriaxone sodium, cephalexin, cephradine, diclofenac

sodium, paracetamol, pefloxacin, and penicillin respectively to the AgNPs-6APA

complex showed no change in the absorption of SPR band. These data suggested that 6-

APA specifically caused quenching of the SPR band, concluding that this analytical

strategy may serve as a potential spectrophotometric probe for assaying 6-APA in the

mixture of other drugs.

Part II

111

4040.00

0.02

0.04

0.06

0.08

0.10

0.12

0.14

Ab

sorb

ance

(un

its)

Wavelength(nm)

ZAgNp+Water

ZAgNp+APA+Water

ZAgNp+APA+Amoxillin

ZAgNp+APA+Aspirin

ZAgNp+APA+Cefaclor

ZAgNp+APA+CefotoximeSodium

ZAgNp+APA+CeftriaxonSodium

ZAgNP+APA+Cephalexin

ZAgNP+APA+Cephradine

ZAgNP+APA+Diclofinic

ZAgNp+APA+Paracetmol

ZAgNP+APA+Pefloxacin

ZAgNp+APA+Penicillin

Figure: 2.33 Effect of interfering drugs on 6-APA quenching

Quenching effect of 6-APA in HumanBlood Plasma

The real samples analysis of our detection system was also explored to check the

quenching sensitivity and selectivity of 4-PyAgNPs for 6-APA in human blood plasma.

Human blood plasma was spiked with 0.05 mM 6-APA solution before use, and the

spiked plasma sample was treated with AgNPs. Fig. 2.34 indicates that the ingredients of

the complex and critically important physiological sample of human plasma did not alter

the photophysical properties of AgNPs, and a considerable magnitude of SPR band

quenching is still observed.

Part II

112

Figure: 2.34 6-APA quenching in Human blood plasma sample

2.2.2.6 Antimicrobial Activity of 4-Py-AgNPs

Thiolated organic moieties are known to have a broad spectrum activities against both

gram positive and gram negative microbes and fungi, therefore these are the active part of

pharmacophores in many developed antibiotic drugs for example cephalosporin,

amoxicillin etc.

In this study we used a complex strain of E. coli ATCC 8739; it is a human pathogen that

is considered as a reason of urinary tract infections, gastroenteritis, and neonatal

meningitis (Croxen and Finlay 2010). To study the antibacterial activity of 4-Py-AgNPs,

we utilized AFM along with the conventional Agar well diffusion method. The minimum

inhibitory concentrations (MIC) were calculated by using zone of inhibition method.

The membrane surface of E. coli is not considered as smooth at nanoscale since it is

made up of inhomogeneous packing of lipopolysaccharide (LPS). The packing of the

Part II

113

adjacent patches is tight, which provides an effective permeability barrier for the E. coli

(Coughlin, Caldwell et al. 1981). Metal ions play a key role in upholding the assembly of

the LPSs and membrane proteins within the outer membrane (Coughlin, Tonsager et al.

1983). This technique is usefully exploited to increase the cell permeability and the

liberation of proteins. This phenomenon is being repeatedly reported in the literature as a

method to attain the liberation of the periplasmic substances of Gram-negative bacteria

can be attained by the depletion of metals from the surface of these organisms (Amro,

Kotra et al. 2000).

Cu(I) accumulation dose for E. coli ATCC 8739 is found to be 25µg/ml to 35µg/ml in

which the bacterial cells digested the Cu(I) for their metabolic system regulation. In

excesses amount of Cu(I) like 40µg/ml and above, accumulation was stated toxic for E.

coli cells. Data is represented in table 2.1.

Table 2.1 Zone of inhibition data at multiple dose of Cu(I) accumulation

CuCl Cuprous chloride

CuCl accumulation dose µg/ml Inhibition

1 -

5 -

10 -

25 -

40 9mm±.1

50 10mm±.1

100 12mm±0.3

200 14mm±0.5

300 15mm±0.5

400 17mm±0.4

500 19mm±0.3

Part II

114

Minimum Inhibitory concentration of 4-Py ligand is calculated from 200µg/ml to

300µg/ml, whereas 4- Py-AgNPs of Para-formyl pyridinium propyl thioacetate ligand has

contained 100µg/ml to 200µg/ml. The MIC of 4- Py-AgNPs as compared to ligand is

found to be two times higher, while the bar graph in Fig. 2.35 represents the data.

Figure: 2.35 MIC of ligand in comparison with ligand stabilized 4- Py-AgNPs

in the presence of Cu(I).

The nanoparticles can easily penetrate the bacterial cell membrane by adhering to

proteins in the cell membrane due to the high affinity of 4- Py-AgNPs owards treated

Cu(I) ions. The pathways involved in bacterial responses to 4- Py-AgNPs are highly

dependent on physicochemical properties of the nanoparticles, and in particular the surface

characteristics. These results haveimportant implications for the regulation and testing of

silver nanoparticles (Carlson, Hussain et al. 2008).

0

5

10

15

20

25

30

5 10 25 50 100 200 300 400 500

Zon

e o

f in

hib

itio

n m

m

MIC µg/ml

Zones of inhibition of Ligand(mm)

Zones of inhibition of AgNPs Ligand(mm)

Part II

115

AFM analysis also confirmed the results of the zone of inhibition assay. Since the MIC of

ligand was around 2 times less as compared to its 4- Py-AgNPs, severe morphological

changes are expected in the nanoparticles treated E. coli. While unconjugated 4- Py-

AgNPs exhibited very poor MIC values in our previous report, hence its role as cytotoxic

agent itself is ruled out (Shah, Ali et al. 2014).

Images of untreated E. coli (control) cells revealed a relatively smooth surface with no

ruptures. The surface of E. coli is found to be smooth and shows no indentations in Fig.

2.36A. While Fig. 2.36B represents the Cu(I) treated E. coli, it can be clearly seen that

Cu(I) treated cellslooks healthier than the control cells. Since at 30 µg/mL (below the

MIC), Cu(I) ions are used in the metabolism of the E. coli.

AFM images were acquired at MIC of sample on E. coli. Only relatively intact cells were

imaged, and the images therefore represent only cells with minimal lytic damage at a

particular sample concentration. In general, the roughness of the outer membrane

increased with the increase in sample concentration, with ligand coated 4- Py-AgNPs

having the most pronounced effect on the surface roughness.

The cultures were then treated with formyl pyridinium thioacetate ligand at a dose of 200

µg/mL, and cell morphology was analyzed via AFM as a function of incubation time.

The sample exhibited rough surfaces after 4 h (Fig. 2.37). While the cellular degradation

increased with time, and after 8 h the cells were significantly degraded. Since the MIC

data suggests that 4- Py-AgNPs more effectively inhibit the growth of E. coli as

compared to pure ligand. The AFM analysis presented an interesting feature in

accordance with this data. 4- Py-AgNPs were found to degrade cells faster than pure

compound, Ag conjugates below MIC that is at 100 µg/mL concentration induced cell

rupture by 4 h (Fig. 2.38A). While at higher incubation time that is after eight hours, the

cells were completely melted and destroyed. Significantly, unconjugated Au and Ag NPs

at even higher dosage did not induce any significant morphological changes even after

long incubation time (i.e.48 h).

Part II

116

Figure: 2.36 A) E. coli Control. B) E. coli after treating Cu(I)

Figure: 2.37 Ligand treated E. coli at A) 4 hours. B) 8 hours

Figure: 2.38 4- Py-AgNPs treated E. coli at A) 4 hours. B) 8 hours

Part II

117

2.2.3 Synthesis of Pyrazinium Thioacetate Gold Nanoparticles Pyr-AuNPs and their

Applications

2.2.3.1 Synthesis of Pyr-AuNPs

Pyrwas dissolved in 10% mixture of methanol to deionized water for preparing 0.1 mM

solution, while 0.1 mM solution of tetrachloroauric (III) acid was prepared in deionized

water only. 1 mL (0.1 mM) of Pyr was added into 10 mL (0.1 mM) of tetrachloroauric

(III) acid and the mixture was stirred for 10 minutes. Then 0.1 mL of freshly prepared 4

mM NaBH4 solution was added in the reaction mixture. The color of the reaction mixture

rapidly changed from colorless to wine red. The resulting mixture was stirred for further

1 hour at room temperature, and was used as reference AuNPs solution throughout this

study. The colloidal AuNPs are stable at room temperature and ordinary atmosphere for

more than 1 month. Solid AuNPs were collected by freeze drying.

2.2.3.2 Characterization of Pyr-AuNPs

350 400 450 500 550 600 6500.00

0.05

0.10

0.15

0.20

0.25

0.30

Abso

rban

ce(u

nits

)

Wavelength(nm)

0.1mMAuNPs

Figure: 2.39 UV-visible spectrum of Pyr-AuNPs

In this study, the AuNPs are synthesized by simple reduction of HAuCl4 via NaBH4 in

the presence of the stabilizing agent (one phase method). It is the most widely used

method for the synthesis of AuNPs due to its robustness and rapidity. The one phase

Part II

118

method is more useful as compared to other common methods because it is simple, and

does not require any special treatment like heating, incubation etc. However, using

NaBH4 for the rapid reduction of metal salt may lose the size selectivity for the formation

of relatively small nanoparticles. Along with compromising size selectivity, the toxicity

of borohydrides restricts its use for the synthesis of nanoparticles for medical application.

But for the purpose of chemosensing, this method works fine.

For optimization of the synthetic procedure, various ratios of Pyr and HAuCl4 were

applied, among which 1 ratio 10 was finally chosen, due to stability and prominent SPR

band absorption of formed AuNPs. The SPR maxima were observed at 520 nm, Fig.2.39

displays the typical absorption spectra of colloidal gold stabilized with thiolated ligand.

Notably, the synthesis process only required 15 min, and no further purification or any

treatment was needed. Further characterization of Pyr-AuNPs was achieved by FT-IR

spectroscopy and AFM which will be discussed later.

2.2.3.3 Photophysical Applications of Pyr-AuNPs

In the present study we developed a simple two steps procedure for the synthesis of 1-(3-

(acetylthio) propyl) pyrazin-1-ium capping agent for the stabilization of AuNP. The

obtained AuNPs were used as sensor for detection and quantification of Fe(III) and

Pd(II). The analytical potential of this nanosensor was evaluated by screening tap water

and blood plasma containing Fe(III) and Pd(II). Our schematic design offers key

advantages over several other metal detecting systems as it is stable and robust. Unlike

particular organic fluorophores used recently for detecting heavy metals, the synthesis of

Pyr is quite simple, while AuNPs can also be prepared rapidly without the need of tedious

separation techniques. The high dispersibility of the AuNP probes in aqueous solution

gives this method an edge over recent reported methods for the sensitive detection of

heavy metal ions in water. It provides rapid sensing of heavy metals like Fe (III) and

palladium in very economical way through UV-visible spectrophotometry, while the

sensitivity and specificity of the system is easily reproducible. Finally our system can be

used potentially for the detection and removal of trace heavy metals from drinking water

and physiological fluids.

Part II

119

2.2.3.3.1 Recognition of Fe (III )with Pyr-AuNPs

400 450 500 550 600 650 700 7500.0

0.1

0.2A

bso

rban

ce(u

nit

s)

Wavelength(nm)

AYNP only

AYNP+Ca(II)

AYNP+Cu(I)

AYNP+Cu(I)

AYNP+Cu(II)

AYNP+Cu(I)

AYNP+Fe(II)

AYNP+Fe(III)

AYNP+Fe(II)

AYNP+K(I)

AYNP+Na(I)

AYNP+Ni(II)

AYNP+Rb(I)

AYNP+Cu(I)

AYNP+Sb(III)

Figure: 2.40 UV-visible spectra of metal salts screening

Because heterocyclic ligands are known to form complexes with Fe(III) ions as in heme,

we explored our pyrazinium stabilized AuNPs for the sensing of Fe(III) ions, through a

mechanism based on the complexation of the Fe(III) ions in solution with the Nitrogen

atom of the pyrazine.

The optical sensitivity of heavy metal ions with respect to prepared nanoparticles was

investigated in 1:1 mixture of 0.1 mM metal salt solutions and AuNPs. The UV–vis

spectra (Fig. 2.40) of AuNPs were recorded after 15 minutes of addition of salt solutions.

A significant decrease in absorption of SPR band was observed within 15 min after the

addition of Fe (III) ions to AuNPs, presumably because of aggregation-induced

quenching. The appearance of a quenched SPR band for Fe (III), indicates that the

prepared AuNPs was sensitive and selective towards Fe(III).

Part II

120

Figure: 2.41 Buffer stability of AuNPs with and without Fe(III)

For the stability of AuNPs in aqueous solution, pH of the media needs to be optimized so

that the background agglomeration will be minimal. The effect of pH on the Pyr-AuNPs

is shown in Fig. 2.41. The pH of colloids is found to be 8.4, the solution is slightly basic

due to free pyrazine groups on the surface of gold nanoparticles. The effect of pH on

SPR band was studied by varying pH from 2–12. It is observed that the variation of pH

did not affect the stability of AuNPs, but due to dilution a decrease in absorption of SPR

band was observed.

We further investigated the effect of pH on the optical responses of the AuNPs to Fe(III).

In the presence of Fe(III) ions (0.1 mM), the colloids showed the highest absorbance at

pH=12, while the absorbance was quenched most at pH=2. Since interaction of the

Fe(III) ions with AuNPs is influenced by pH of the solution so investigation of pH is an

important step in chemosensing. The best pH for interaction of Fe(III) ions with AuNPs

was achieved around 2-3. Such a profile indicates, that for pH values lower and higher

than 2-3 in accumulation step, the amount of accumulated Fe(III) ions on AuNPs was

decreased, due to H+ competition for functional groups of the surface, and OH−for Fe(III)

in solution.

0 0.02 0.04 0.06 0.08

0.1 0.12 0.14 0.16 0.18

2,3 4,5 6,7 8,9 10,11 11,12

Ab

sorb

ance

(un

its)

pH(units)

AYNP +FeCl3

AYNP only

Part II

121

2.2.3.3.3 FT-IR spectral analysis

Figure: 2.42 Comparative FT-IR spectra of Pyr-AuNPs before and after mixing of Fe(III)

The FT-IR spectrum of ligand indicates the presence of two peaks of interest as, a weak

band of C-S at 617, and thioester’s carbonyl at 1635 cm-1, while the rest of characteristics

peaks of the chemical structure of Pyr are present in the spectrum. The formation of Pyr-

AuNPs is ascribed to the disappearance of the thioester peak, which implies that the

stabilizing head group of the ligand must be –SH. Fig. 2.42 represents the FT-IR

spectrum of the typical thiolated AuNPs, where strong and broad band around 3500 is

associated with the water in the medium. FT-IR spectra of AuNPs before and after

mixinng with Fe(III) is shown in Fig. 2.42, and on the basis of the observation that no

change is occurred between the two spectra, it can be safely concluded that the

mechanism of SPR quenching is not related to any kind of chemical transformation or

detachment of the ligand from AuNPs.

0

10

20

30

40

50

350 850 1350 1850 2350 2850 3350 3850

% T

ran

smit

tan

ce

Wave number(1/cm)

AuNPs+Fe

AuNPs

Part II

122

2.2.3.3.4 Morphology of AuNPs

Figure: 2.43 AFM images of AuNPs alone and of AuNPs treated with Fe(III)

Part II

123

The morphological information of the AuNPs is obtained by AFM, and is displayed in

Fig. 2.43. It showed the presence of polydispersed and spherical particles with in the size

range of 15-100 nm. Hence the applied reaction conditions do not give rise to size

selectivity for the synthesis of AuNPs. The AFM images reveals that the AuNPs were

initially small, and tended to aggregate in the presence of Fe(III) ions, resulting in the

formation of huge complexes of size in the range of 100-1000 nm. However, the obtained

behavior of AuNPs is very attractive for its application in optical sensors, since the shape

of the nanoparticles is related to its optical properties and it justifies the instant decrease

in SPR band of AuNPs was occurred due to aggregation.

2.2.3.3.5 Quantitative relationship between Nanosensor and Fe(III)

Figure: 2.44 Concentration study with the calibration curve

On the basis of FT-IR spectral analysis, it can be safely assumed that, when ferric

chloride is added to the AuNPs, Fe(III) ions interact with the Nitrogen of pyrazine which

are free at the surface of the AuNPs. So, to demonstrate the sensitivity of our nanosensor

for the quantitation of Fe(III) ions by the mechanism mentioned above, UV-visible

Part II

124

spectra of AuNPs in the presence of various concentrations of Fe(III) was recorded (Fig.

2.44). It shows that there is a decrease in absorbance on increasing the concentration of

Fe(III) ions. The data exhibited a good linear correlation to Fe(III) concentration in the

range from 1 to 100 μM. The regression constant (R2) is 0.9813, while the limit of

detection (LOD) and the limit of quantification (LOQ) are found to be 4.3 μM and 13.19

μM respectively. Since, The LOD of our detection system is lower than the maximum

level (5.4 μM) of Fe(III) ions permitted in drinking water by the US Environmental

Protection Agency, we hope that this method can turn out as a cheap alternative for the

detection of Fe(III) ions in drinking water.

2.2.3.3.6 Selective interaction of Pyr-AuNPs with Fe(III) in the presence of competing

metal ions

5380.00

0.02

0.04

0.06

0.08

0.10

Ab

sorb

ance

(un

its)

Wavelength(nm)

AYNP

AYNP+Fe(III)

AYNP+Fe(III)+K(I)

AYNP+Fe(III)+Na(I)

AYNP+Fe(III)+Ni(II)

AYNP+Fe(III)+Cu(II)

AYNP+Fe(III)+Cu(I)

AYNP+Fe(III)+Cu(I)

AYNP+Fe(III)+Na(I)

AYNP+Fe(III)+Sb(III)

Figure: 2.45 Effect of interfering metal salts on the Fe(II) sensing

Since a practical detection system must possess a high selectivity and rapid response time

to differentiate and recognize the target among potentially competing entities. Therefore,

to test the specificity of the developed optical probe for Fe(III) ions, the nanosensor

discriminates Fe(III) in the presence of other competing metal ions. The concentration of

Part II

125

the interfering salts was kept same as that of Fe(III) (0.1 mM). As shown in Fig.2.45, in

the presence of Fe(III) ions, no response is observed toward K(I), Na(I), Ni(II), Cu(II),

Cu(I), and Sb(III). Small shift in the plasmon band energy to longer wavelength is

observed in the presence of K(I) and Cu(I), however, the responses are negligible

compared with that of Fe(III). Hence Fig. 2.45 establishes that the chemosensor has

relatively desirable selectivity towards Fe(III).

Part II

126

400 500 600 700 800

0.0

0.1

0.2

Ab

sorb

ance

(un

its)

Wavelength(nm)

Fe(III)+tap water

Fe(III)+tap water+AYNPs

AYNPs

Tap water

A

400 500 600 700 8000.0

0.1

0.2

Ab

sorb

ance

(un

its)

Wavelength(nm)

AYNPs

Plasma+Fe(III)+AYNPs

Plasma+Fe(III)

Plasma

B

Figure: 2.46 Fe(III) sensing in drinking water and human blood plasma

Part II

127

The analytical potential of the nanosensor was exploited through analysis of Fe(III) in

real samples. For this purpose, we selected human blood plasma taken from healthy

human volunteer, which was used after standard pretreatment protocol, and tap water

taken from Karachi University. Both of these samples were spiked with the 0.1 mM

FeCl3 solution before use. We observed the Fe(III) dependent quenching in the SPR band

of AuNPs is not affected by the complex analyte samples and the nanosensor response is

distinctive towards Fe(III) (Fig. 2.46).

2.2.3.3.8 UV-visible Spectrophotometric Detection of Pd(II)via Pyr-AuNPs

A series of eighteen salts of 0.1mM concentration were mixed with Pyr-AuNPs in 1:1

ratio. Interestingly we observed the disappearance of color of AuNPs with Pd(II) (Fig.

2.47A). All other salt solutions showed no visible changes at all on AuNPs color such as

Ag(I), Ba(II), Ca(II), Cd(II), Co(I), Cr(III), Cu(I) , Cu(II), Sb(III), Fe(II), Hg(II), K(I),

Mg(II), Na(I), Ni(II), Pb(II), and Rb(I). The significant color change can be attributed to

the destabilization of AuNPs induced by Pd(II). Further spectral analysis results of

mixture of metal ions and Pyr-AuNPs screening (Fig. 2.47) demonstrates that Pd(II)

induced aggregation of AuNPs, hence the surface plasmon resonane (SPR) band is

completely quenched. These results also reveals that Pyr-AuNPs can serve as a selective

“naked-eye” off–on probe for Pd(II) recognition.

Part II

128

400 500 600 7000.0

0.1

0.2Ab

sorb

ance

(uni

ts)

Wavelength(nm)

AYNP+Ag(I)

AYNP+Ba(II)

AYNP+Ca(II)

AYNP+Cd(II)

AYNP+Co(I)

AYNP+Cr(III)

AYNP+Cu(I)

AYNP+Cu(II)

AYNP+Sb(III)

AYNP+Fe(II)

AYNP+Hg(II)

AYNP+K(I)

AYNP+Mg(II)

AYNP+Na(I)

AYNP+Ni(II)

AYNP+Pb(II)

AYNP+Rb(I)

AYNP+Sb(III)

AYNP+Pd(II)

AYNP only

B

Figure: 2.47A) Pictorial display of colorimetric sensing of Pd(II), B) UV-visible

spectrum of salt screening

2.2.3.3.9 Effect of pH on AuNPs Stability and Pd(II) Sensing

For the stability of AuNPs in aqueous solution, pH of the media needs to be optimized so

that the background agglomeration become minimal. The bar graph diagram (Fig. 2.48)

shows the effect of pH on the Pyr-AuNPs. The pH of colloids is found to be 8.4, the

solution is slightly basic due to free pyrazine groups on the surface of AuNPs. The effect

of pH on SPR band was studied by varying pH from 2–12. It is observed that the

variation of pH did not affect the stability of AuNPs, but due to dilution a little decrease

in absorption of SPR band was observed.

The pH of the medium has great influence on the optical response of the complexation

reaction, thus an optimum pH level is essential to develop maximum interaction between

AuNPs and Pd(II) complex. The comparison in intensity of SPR band of the AuNPs-

Pd(II) mixture proves a unique feature of this detection system that the quenching is

independent of the pH change (Fig. 2.48). Since pH variation yields no significant change

on the absorption behaviour of Pyr-AuNPs so our system can be used at any pH.

Part II

129

Figure: 2.48 pH stability of AuNPs with and without Pd(II)

2.2.3.3.10 Morphological investigation of Pyr-AuNPs and its adduct with Pd(II)

The morphology of the AuNPs is investigated by AFM images, the results and analysis

are displayed in Fig.2.49. Fig.2.49a shows the presence of polydispersed and spherical

particles with in the size range of 20-90 nm. Since the adopted reaction conditions do not

give rise to size selectivity for the synthesis of AuNPs, so nanoparticles in wide range of

size were obtained as revealed by AFM analysis. The AFM images in Fig. 2.49b reveals

that the AuNPs were initially small, and were prone to aggregation in the presence of

Pd(II) ions, resulting in the formation of huge complexes of upto 1 µm scale. These

results are in excellent agreement with the UV-visible spectrophotometric data, and

thebehaviour of AuNPs is turned out to be attractive for its application in optical sensing.

Since the size and shape of the metal nanoparticles are related to their optical properties,

it justifies the instant decrease in SPR band of AuNPs which occurred due to aggregation.

Part II

130

Figure: 2.49 A) AFM image of AuNPs, B) AFM image of AuNPs after mixing

with Pd(II)

Part II

131

2.2.3.3.11 FT-IR Spectral Analysis

Since Pd(II) species are known to form complexes with nitrogenated ligands, FT-IR

spectra of AuNPs before and after treatment with Pd(II) suggest the interaction of Pd(II)

with the pyrazine moiety as a result of which C-N band is shifted from 1115 to 1250 cm-

1a region of much higher energy.

2.2.3.3.12 Kinetic evaluation of supramolecular interaction between Pyr-AuNPs and

Pd(II)

Kinetics associated to supramolecular interaction of Pyr-AuNPs and Pd(II) was

investigated by varying the concentration of Pd(II) while keeping constant the

concentration of nanoparticles during mixing. Fig. 2.50a represents the spectral results,

while inset shows the calibration curve which indicated that the data strictly followed

beer’s law and exhibited a good linear correlation to Pd(II) concentration in the range

from 1 to 80 μM with the regression constant (R2) equal to 0.9402. The limit of detection

(LOD), and limit of quantification (LOQ) are found to be 4.23 μM and 12.83 μM.

Moreover, to estimate the binding stoichiometry between AuNPs and Pd(II) Job's plot

experiment was performed. Absorption spectra were recorded by varying the mole ratio

of Pd(II) to AuNPs. Fig. 2.50b displays that the stable AuNPs-Pd(II) is formed at 0.4, 0.6

mole ratios of Pd(II) and AuNPs respectively.

400 450 500 550 600 650 700 750 800

0.00

0.05

0.10

0.15

0.20 y = 0.0073x - 0.0204R² = 0.9402

0

0.02

0.04

0.06

0.08

0.1

0.12

Abso

rban

ce(u

nits

)

Wavelength(nm)

.01uM

.05uM

.1uM

.5uM

1uM

5uM

10uM

20uM

30uM

40uM

50uM

60uM

70uM

80uM

90uM

100uM

A

Part II

132

Figure: 2.50 A) UV-visible spectral profile of the concentration study, inset represents

calibration curve, B) Job’s plot

2.2.3.3.13 Selective interaction of Pyr-AuNPs with Pd(II)in presence ofCompeting

Metals Ions

Since a practical metal chemosensor must possess a high selectivity and rapid response

time to differentiate and recognize the target metal ion among potentially competing

entities, the Pd(II) specificity was assessed by competitive experiments. The absorbance

changes of AuNPs were measured by contacting 1 equivalent of Pd(II) in the presence of

1 equivalent of other interfering metal ions including Cu(I) , Cu(II), Na(I), K(I), Rb(I),

Ni(II), and Sb (III). As shown in Fig. 2.51, the tested metal ions experienced no

interference between the interaction of Pd(II) and Pyr-AuNPs. Hence this specific nano

sensor can be used to recognize Pd(II) ions in a mixture of several other metal ions

without interfering from competing metal ions.

Part II

133

5400.00

0.02

0.04

0.06

0.08

0.10A

bso

rban

ce(u

nit

s)

Wavelength(nm)

AYNP

AYNP+Pd(II)

AYNP+Pd(II)+Cu(II)

AYNP+Pd(II)+Cu(I)

AYNP+Pd(II)+Na(I)

AYNP+Pd(II)+K(I)

AYNP+Pd(II)+Rb(I)

AYNP+Pd(II)+Ni(II)

AYNP+Pd(II)+Sb(III)

Figure: 2.51 Effect of interfering metal ions on Pd(III) sensing

2.2.3.3.14 Detection of Pd(II) in Tap Water and Human Blood Plasma

The analytical application of Pyr-AuNPs to detect Pd(II) in real samples like tap water

and Human blood plasma was also explored. For this purpose, we used human blood

plasma obtained from healthy human volunteer, which was used after standard

pretreatment protocol, and tap water taken from Karachi University. Fig. 2.52a indicates

that a significant magnitude of SPR band quenching is still observed in AuNPs with 0.1

mM Pd(II) spiked tap water. While in case of blood plasma a slight blue shift is observed

with less quenching of SPR band absorbance than 0.1 mM Pd(II) aqueous solution (Fig.

2.52B). These results suggest that our system can be effectively used to detect Pd(II) in

water and blood testing. It is also concluded from the study that the ingredients of tap

water and Human blood plasma did not interfere in the detection process.

Part II

134

Figure: 2.52 UV-visible spectra of Pyr-AuNPs after mixing with A) tap water, B)

Human blood plasma spiked with Pd(II)

Part II

135

2.2.4 A New Pefloxacin Chemosensor Based on Thioacetate Functionalized DMAP

coated Gold Nanoparticles

We developed a synthetic route for the facile synthesis of dimethyl aminopyridine based

thiolated capping agent. The capping agent was used to coat AuNPs which effectively

recognized Pefloxacine (Pef) in aqueous solution. Importantly this protocol provides an

easy, relatively rapid, economical and sensitive method for detection of Pef in water and

in human blood plasma samples. We hope that this method can be utilized for the

recognition and control of this antibiotic in drinking water as well as in important

physiological fluids.

2.2.4.1 Synthesis of Dimethylaminopyridinium Propylthioacetate (D2) Stabilized Gold

Nanoparticles (D2-AuNPs)

1 mL of 0.1 mM solution of D2 was added in a shaking solution of 10 mL of 0.1

Chloroauric acid trihydrate mM . After 15 minutes, 0.1 mL of freshly prepared 4 mM

NaBH4 solution was added in the reaction mixture. The colorless reaction mixture turned

into clear wine red instantly after mixing of NaBH4 solution, and the reaction mixture

was kept on shaking for 2 more hours. The resulting solution was subjected to UV-visible

spectrophotometry and AFM imaging.

D2-AuNPs were synthesized via one phase method by NaBH4 reduction. Various ratios

of Gold salt and D2 were used, and 10:1 with respect to Gold salt and D2 was selected

based on the prominent peak of AuNPs surface plasmon band at 520 nm. UV-visible

spectrum of the colloids was recorded after 2 and 4 hours. Since there was no difference

in absorbance intensity at longer time, the formation of nanoparticles was achieved at 2

hours shaking. However an increase in color intensity of nanoparticles solution was

observed after 3 days, and the UV-visible spectrum indicates the formation of larger

aggregates at prolonged time (Fig 2.52A). Fig. 2.52b reveals the morphology of the

nanoparticles, they were found to be spherical and poly dispersed in size. While the

particles size lie in the range of 15-45 nm (Fig. 2.52C).

Part II

136

Part II

137

Figure: 2.53 A) UV-Visible spectrum of the AuNPs stabilized with D2, B) AFM image

of the D2 capped AuNPs, C) Size distribution histogram of D2-AuNPs

2.2.4.2 Stability of D2-AuNPs

Thermal stability of AuNPs was tested at 100 oC for 1 hour. Only a slight increase in

intensity of SPR was observed, but no change was appeared in the dispersety of the

solution, even on heating for longer time (Fig.2.53A). This effect in the SPR band

maxima is described by electron dephasing mechanism in the literature (Link and El-

Sayed 1999). Furthermore, chloride promoted aggregation of the nanoparticles was also

studied. Fig. 2.53b shows that the D2-AuNPswere stable over the wide range of 0.01 mM

to 10 mM concentration of sodium chloride (NaCl) with the decrease in intensity of SPR

band (Bae, Nam et al. 2002). At NaCl concentration of 50 mM or above only, the SPR

band broadening and visible aggregation of the AgNPs was observed.

Part II

138

Figure: 2.54 A) Effect of heating on SPB, B) Aggregation of D2-AuNPs induced by

various concentration of NaCl

Part II

139

2.2.4.3 Exploration of Chemosensing Potential of D2-AuNPs

The recognition behavior of fully characterized D2-AuNPswas assessed for various drugs

by mixing 1:1 v/v ratio of 0.1 mM drugs solutions and AuNPs. The UV–vis spectra of

AuNPs were recorded instantly after addition of drug solutions. The effect of tested drugs

such as Pefloxacin mesylate, Paracetamol, Penicillin, Amoxicillin, Cefaclor, Cefotaxime

sodium, Ceftriaxone sodium, Cephalexime, and Cephradine on absorption intensity was

investigated and plotted in Fig. 2.55. All drugs induced the enhancement in the

absorbance intensity of D2-AuNPs, but Pefloxacin mesylate resulted in the hyperchromic

shift, while a new peak around 700 nm is appeared. The red shifted band was presumably

observed due to the induced aggregation of AuNPs. The substantially large difference

between rest of the drugs and Pefloxacin mesylate added absorption behaviour indicates

some chemical or physical interactions between the D2-AuNPsand this drug. Hence,

these results of spectrophotometric drugs screening suggest that this nano probe can be

exploited as a selective sensor for the detection of Pef.

Figure: 2.55 UV-Visible spectral profile generated after mixing drugs with D2-AuNPs

Part II

140

2.2.4.4 Effect of the pH on the Stability of D2-AuNPs and Pef Response

The pH of colloids is found to be in the range of 4-5, the solution is slightly acidic in

nature due to cationic protonated form. The synthesized D2-AuNPs was found to be

stable in both basic media, as well as acidic media. While at neutral pH, the colloids were

prone to aggregation and resulted in the peak broadening of the SPR band.

Since the pH of the medium has a great impact on the optical response of the

complexation reaction, an optimum pH is thus essential to develop for the interaction of

AuNPs-Pef complex. While a practical sensor needs to be smart enough to remain

effective in a wide range of pH. A study of the pH in the range of 2–12 with the

optimized conditions for interaction of nanoparticles with the drug is shown in Fig. 2.56.

The absorbance of SPR band of the D2-AuNPs-Pef mixture shows the maxima at pH 10-

11, and before and after which it gradually decreases. On the basis of these results it can

be safely concluded that the drug and nanoparticles interaction is independent of the pH

change, and our system does not rely on pH of media.

Figure: 2.56 Effect of pH on the interaction of D2-AuNPs with Pef

Part II

141

2.2.4.5 FT-IR Spectral Analysis

To have a deeper understanding of the mechanism of nanoparticles formation, a

comparison between FT-IR spectrum of D2, and D2-AuNPsis studied. Results suggest

that thioester functionality is cleaved and Gold nanoparticle surface is stabilized with

thiol group since the thioester peak at 1716 1/cm in D2 spectrum is diminished after the

formation of AuNPs, while the rest of the characteristics peak for DMAP were all

present. We also recorded the FT-IR spectra of Pef, which showed O-H stretch at 3427,

N-CH3 at 2646, carboxyl carbonyl at 1716, C-O at 1627, C-F at 1091 1/cm. While

sulfoxide asymmetric and symmetric stretches were observed at 1197 and 1051 1/cm

(Dorofeev 2004). Interestingly, in a mixture of D2-AuNPsand Pef, the acid carbonyl

peak which appeared at 1716 1/cm in the drug was absent, and a new peak at 1583 1/cm

appeared. This peak at 1583 1/cm is attributed to the carboxylate moiety, which is

responsible for the accumulation of Pef with AuNPs via hydrogen bonding.

2.2.4.6 AFM investigation of the interaction between D2-AuNPs and Pef

AFM image of the 1:1 mixture of D2-AuNPs and Pef revealed that the nanoparticles are

prone to agglomeration upon interaction with the drug. Fig.2.57 depicts the presence of

larger aggregates ranging in 50-150 nm size. These results are in agreement with that of

UV-visible and FT-IR spectra, and it can be deduced that the drug-nanoparticle

interaction is based on aggregation induced mechanism.

Part II

142

Figure: 2.57 AFM image of the D2-AuNPs -Pef 1:1 mixture

2.2.4.7 Analytical Performance of the Pef Nanosensor

The analytical performance of Pef chemosensor is demonstrated with the help of

calibration curve. The absorption titration of Pef to measure the sensitive optical response

of D2-AuNPsat 700 nm (Fig.2.58) strictly followed beer’s law and exhibited a good

linear correlation in the range from 0.1 to 90 μM with the regression constant (R2) equal

to 0.945, and the limit of detection (LOD) 10.9 μM. Moreover, to estimate the binding

stoichiometry between stable D2-AuNPs-Pef complex, Job's plot experiment was

performed. Absorption spectra were recorded by varying the mole ratio of Pef to D2-

AuNPs. The minimum on the plot between absorbance and mole fraction appeared at 0.5,

which correspond to the 1:1 stoichiometric relation between the stable D2-AuNPs-Pef

complexes.

Part II

143

Part II

144

Figure: 2.58 A) Concentration study of Pef with D2-AuNPs, B) Calibration

curve at 700 nm C) Job’s plot for binding ratio

2.2.4.8 Interference Study

Since it is essential for a practical detection system that it can specifically target the

binding specie in an environment where other competing entities are present. Therefore

to test the specificity of this nanosensor, we evaluated the response of optimized Pef

recognition strategy in the presence of other interfering drugs. All the drugs were used in

1:1 (v/v) against Pef, and the UV-Visible spectra of the mixtures were recorded which are

presented in Fig. 2.59. It shows that the addition of competing drugs such as 6-amino

penicillanic acid, amoxicillin, aspirin, cefaclor, diclofenac sodium, flurbiprofen,

paracetamol, and penicillin respectively to the D2-AuNPs-pef mixture showed no

significant change in the absorption intensity of 700 nm band. These results conclude that

this analytical strategy may serve as a potential spectrophotometric probe for assaying

Pef in the mixture of other drugs.

Part II

145

Figure: 2.59 Effect of competing drugs on Pef response towards nanosensor

2.2.4.9 Analysis of Pef in Real Samples

The utility of the optimized Pef detection system was explored to check the sensitivity

and selectivity of optical response in real and complex samples. We tested the efficiency

of this Pef chemosensor in the analysis of tap water and human blood plasma. Human

blood plasma was collected from healthy human volunteer, which was used after standard

pretreatment protocol, while tap water was taken from Karachi University. Both samples

were spiked with 0.1 mM concentration of pef before analysis. Fig. 2.60A indicates that

the distinctive pef recognition signal is still observed in D2-AuNPs with 0.1 mM Pef

spiked tap water. While in case of blood plasma only a hyperchromic shift without the

700 nm band is observed (Fig. 2.60B). These results suggest that our system can be

effectively used to detect Pef in drinking water treatment and blood testing.

Part II

146

Figure: 2.60 UV-visible spectra of Pef recognition in (A) tap water (B)

Human blood plasma

Part II

147

2.3 EXPERIMENTAL

2.3.1 Materials

All the chemicals (Solvents and reagents) used in the synthesis of ligand were of

analytical grade and used as provided. Oven dried glassware were used, washed with

aqua regia and rinsed by deionized water. Freshly prepared solutions were used

throughout synthesis and analysis. 4-Pyridinecarboxaldehyde, Pyrazine, and Dimethyl

aminopyridine were purchased from Tokyo chemicals industry (TCI, China). Dibromo

propane and potassium thioacetate were purchased from Alfa-Aesar (MA, USA).

Chloroauric acid trihydrate, Triphenylphosphine gold (I) chloride, Borane tert-butylamine

complex (BTBC), and all the bisphosphines were purchased from Sigma-Aldrich (St.

Louis, USA). Other chemicals were purchased from local chemical suppliers.

2.3.2 Instruments

UV-Visible spectrophotometer used for characterization of nanoparticles was Thermo

Scientific Evolution 300 spectrophotometer, operated at 2 nm resolution with a quartz

cuvette of 1cm path length. The spectra were reordered in range 200-800 nm. The FT-IR

spectra were recorded with Bruker-Vector-22, in diffuse reflectance modewith2 cm-1

resolution. Sample preparation was consist of mixing test sample separately with KBr in

1:1 ratio to form the KBr disk. The spectra were recorded between 400 and 4000 cm-1.

Oakton, Eutech model-510 pH-meter having glass-electrode and Ag/AgCl electrode was

used as a reference in pH measurements. Morphological analysis of synthesized nano

particles was carried out by Atomic Force Microscope (AFM) Agilent-5500 operated in

tapping-mode. Sample preparation for AFM consist of sorption of nanoparticles on

freshly cleaved mica wafer surface from a drop for 30 minutes and was then air dried

under controlled environment. AFM was operated in tapping-mode having triangular

shape silicon-nitride cantilever made by Veeco model-MLCT-AUHW with 0.01-0.01N/m

nominal value of spring-constant.

Part II

148

2.3.3 Synthesis of Bisphosphines Coated Gold Nanoclusters

In a typical synthesis, AuPPh3Cl was dissolved in solvent of choice to reach a

concentration of 10-3 M. The desired bisphosphine ligand was then added in equal

molarity and solution was stirred until completely dissolved. Lastly, BTBC is added at a

concentration of 510-3 M and stirring was continued for about 30 min, under Ar gas.

The solution was turned orange and displayed absorption maxima at around 400 nm.

2.3.4 Synthesis of 1-(3-(acetylthio)propyl)-4-formylpyridin-1-ium (4-Py) Coated

Silver Nanoparticles (4-Py-AgNPs)

1 mL of 0.1 mM solution of 4-Py was added in a shaking solution of 10 mL of 0.1 mM

silver nitrate. After 15 minutes, 0.1 mL of freshly prepared 4 mM NaBH4 solution was

added in the reaction mixture. The colorless reaction mixture turned yellow-brown

instantly after addition of NaBH4 solution, but the solution was stirred further for 2 hours.

The AgNPs colloidal dispersions were centrifuged for 20 min till all nanoparticles

precipitated at 10,000 rpm to separate the potential large aggregates and excess unbound

ligand from the broth followed by redispersion in deionized water.

2.3.5 Synthesis of 1-(3-(acetylthio)propyl)pyrazin-1-ium (Pyr) Coated Gold

Nanoparticles (Pyr-AuNPs)

Pyr was dissolved in 10% mixture of methanol and deionized water to prepare 0.1 mM

solution, while 0.1 mM solution of tetrachloroauric (III) acid was prepared in deionized

water only. 1 mL (0.1 mM) of Pyr solution was added into 10 mL (0.1 mM) of

tetrachloroauric (III) acid and the mixture was stirred. After 10 minutes 0.1 mL of freshly

prepared 4 mM NaBH4 solution was added in the reaction mixture. The colour of the

reaction mixture rapidly changed from colourless to wine red, stirring was continued for

2 hours. The colloidal AuNPs are found stable at room temperature and ordinary

atmosphere for more than 1 month. Solid AuNPs were collected by freeze drying.

Part II

149

2.3.6 Synthesis of 1-(3-(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium (D2)

Coated Gold Nanoparticles (D2-AuNPs)

1 mL of 0.1 mM solution of D2 was added in a shaking solution of 0.1 Chloroauric acid

trihydrate mM (10 mL). After 15 minutes, 0.1 mL of freshly prepared NaBH4 solution (4

mM) was added in the reaction mixture. The colourless reaction mixture was turned into

clear wine red solution instantly after addition of NaBH4 solution, and the reaction

mixture was kept on shaking for 2 more hours. The resulting solution was subjected to

UV-visible spectrophotometry and AFM analysis.

2.3.7 Synthesis of Chlorhexidine (CHX) Coated Gold Nanoparticles (CHX-AuNPs)

10 mL, of AuCl4 (0.5 mM) solution is added into the 1 mL CHX solution (0.5 mM) and

the mixture was allowed to stir for 10 minutes. After 10 minutes 30 microlitres, sodium

borohydride (4mM) was added as reducing agent. The intense deep red color

characteristics of colloidal gold were obtained instantly on addition of borohydride. The

solution was stirred for two more hours. This solution was subjected to UV-Visible

spectrophotometry and Atomic Force Microscopy in order to confirm the successful

formation of gold nanoparticles.

2.3.8 Synthesis of Neomycin (Neo) Coated Silver Nanoparticles (Neo-AgNPs)

10 mL, Silver nitrate aqueous solution (0.1mM) is added into the 2 mL, Neo aqueous

solution (0.1 mM) and the mixture was allowed to stir for 10 minutes. Then 0.5 mL,

sodium hydroxide solution (30mM) was added as mild reducing agent. Shaking was

continued for six hours, after which the solution was subjected to characterization as

mentioned above.

2.3.9 Synthesis of Guanidinobutyric Acid (GBA) Coated Silver Nanoparticles (Neo-

AgNPs)

1 mL, Silver nitrate aqueous solution (0.1mM) is added into the 1 mL, GBA solution (0.1

mM) and the mixture was allowed to stirr. After 10 minutes 1.5 mL, sodium hydroxide

Part II

150

solution (30mM) was added as mild reducing agent, and the reaction was further stirred

for 6 hours.

2.3.10 Cell Incubation

Cytotoxic T lymphocyte (CTLL-2) from (ECACC, ATCC) for AFM based study were

used. For this purpose, the CTLL-2 (ATCC® TIB-214™) cells was cultured in ATCC-

formulated complete RPMI 1640 ((1mM Sodium pyrovate, 2M HEPES and 10% FBS,

10% T-STIM & Con A, 1, 1% L-glutamine and Non-Essential Amino Acids) culturing

media in 75 cm2 flasks, and kept in 5% CO2 incubator at 37oC. After complete

confluences about 5×104 cell/well (in 96 well flat bottom plate corning) of cell line were

used to conduct the experiments.

2.3.11 Cell Treatment

The cell permeability of the 4Py-AgNPs on T-cells was evaluated by incubating 4Py-

AgNPs at different doses with T-cells for 30 min at 37 oC in 5% CO2 incubator. After

incubation the cells were centrifuged at 1300 rpm for 4 min and the cell pellet were

washed twice with 1X, PBS (Phosphate buffer saline) buffer to remove the unbound test

sample. Then the treated cells were incubated for 20 min with multiple concentration of

Cu+ solution at 37 oC in 5% CO2 incubator followed by AFM microscopy.

2.3.12 MTT Based Cytotoxicity Assay

Cytotoxicity of test samples were evaluated in 96-well flat-bottomed micro plates by

using the standard MTT (3-[4, 5-dimethylthiazole-2-yl]-2, 5-diphenyl-tetrazolium

bromide) colorimetric assay. For this purpose, the lymphocytes cells were cultured in

their respective media RPMI 1640 in 75 cm2 flasks, and kept in 5% CO2 incubator at 37 oC. Cell culture with the concentration of 5x104cells/ml was prepared and introduced

(100 µL/well) into 96-well plates. After overnight incubation, medium was removed and

200 µL of fresh medium was added with different concentrations of test sample (30,

50,100 µM). After 48 hrs, 200 µL MTT (0.5 mg/ml) was added to each well and

incubated further for 4 hrs. Subsequently, 100µL of DMSO was added to each well. The

extent of MTT reduction to formazan within cells was calculated by measuring the

Part II

151

absorbance at 540 nm, using a micro plate reader (Spectra Max plus, Molecular Devices,

CA, USA). The percent inhibition was calculated by using the following formula:

Inhibition (%) = [1 - (treated/control)] × 100

Viability (%) = 100 – inhibition (%)

2.3.13 Minimum Inhibitory Concentration by Agar Well Diffusion Method

To calculate minimum inhibitory concentration (MIC), the agar well diffusion method

was employed. Estimation of Minimum inhibitory concentration for 4-py ligand was

measured with or without Ag nanoparticle. In brief, Mueller Hinton agar was used as

medium to grow a lawn of E.coli ATCC 8739at the concentration of 106 cells in one ml.

Separately, Mueller Hinton agar plates carried Cu(I) was made for 4-Py-AgNPs.

Duplicate dilutions were used to calculate minimum inhibition zones. The 60mm well

was made by using a borer. The 100 µg ml¯1 stock solution of 4-Py was used to avoid

nonspecific merged zones of inhibition. In each well different amounts of various

concentrations ranging from 80-100µg ml¯1were added. The plates were incubated at

room temperature for 2 hours to allow the diffusion process to take place before it was

incubated for 24-48 hours at 37 ºC ±1. The zones of inhibition were measured by using a

millimeter scale.

2.3.14 Atomic Force Microscopy Imaging

E. coliATCC 8739 was developed on tryptic soy agar (Oxoid UK) at 37 oC for 24 h in

static conditions, which were marked as stock E. coli culture. A 10 µL droplet of each

test sample was applied onto a poly-L-lysine (PLL) coated glass slide and allowed to

stand at 25 °C for drying, in the meanwhile, a freshly incubated culture of E. coli on

tryptic soy agar (Oxoid UK) was diluted in distilled water to make 106 cfu of E. coli and

10 µL droplets of this solution were transferred onto a freshly cleaved mica surface and

left to dry. Control samples were not treated with the ligand. After deposition, the

samples were rinsed several times with Milli-Q water, and air-dried at 25 °C. The dried

sample was characterized by atomic force microscopy to check its size and morphology.

The test samples were added into vials of distilled water containing 106 cfu of E. coli

Part II

152

bacteria and incubated for 2 h at 37 oC. All experiments were performed with duplicate

cultures for each sample.

The same procedure was applied for 4, and 8 h to check the degradation of E. coli cells.

4-Py-AgNPs dose of 100 µg were treated with 106 cfu of E. coli for 4 and 8 h and were

characterized by atomic force microscopy to check the cell changes and note the effects

of these AgNPs. Atomic force microscope was used (AFM, Agilent Technologies 5500,

USA) in the ACAFM mode. We used a high frequency Si cantilever of 125 µm length,

force constant 42 N m-1 and resonance frequency 330 kHz. All the E. coli samples were

prepared and analyzed under same condition.

Part II

153

REFERENCES

Abdalla, M. A. (1991). "Selective Spectrophotometric Determination of Some

Cephalosporins in Pharmaceutical Formulations." Analytical Letters 24(1): 55-67.

Ackerson, C. J., P. D. Jadzinsky, et al. (2005). "Thiolate Ligands for Synthesis of Water-

Soluble Gold Clusters." Journal of the American Chemical Society 127(18):

6550-6551.

Ajlec, R. and J. Štupar (1989). "Determination of Fe(III) species in wine by ion-exchange

chromatography-flame atomic absorption spectrometry." Analyst 114(2): 137-

142.

Ali, M. and A. Mohammad (1994). "Polarogaphic determination of cephradine in

aqueous and biological media." Bioelectrochemistry and Bioenergetics 33(2):

201-204.

Amro, N. A., L. P. Kotra, et al. (2000). "High-resolution atomic force microscopy studies

of the Escherichia coli outer membrane: structural basis for permeability."

Langmuir 16(6): 2789-2796.

Andrews, N. C. (2000). "Fe(III) metabolism: Fe(III) deficiency and Fe(III) overload."

Annual Review of Genomics and Human Genetics 1(1): 75-98.

Annie Ho, J.-a., H.-C. Chang, et al. (2012). "DOPA-mediated reduction allows the facile

synthesis of fluorescent gold nanoclusters for use as sensing probes for ferric

ions." Analytical Chemistry 84(7): 3246-3253.

Ateeq, M., M. R. Shah, et al. (2014). "Green synthesis and molecular recognition ability

of patuletin coated gold nanoparticles." Biosensors and Bioelectronics.

Badawy, A. M. E., T. P. Luxton, et al. (2010). "Impact of environmental conditions (pH,

ionic strength, and electrolyte type) on the surface charge and aggregation of

silver nanoparticles suspensions." Environmental Science & Technology 44(4):

1260-1266.

Part II

154

Bae, C. H., S. H. Nam, et al. (2002). "Formation of silver nanoparticles by laser ablation

of a silver target in NaCl solution." Applied Surface Science 197–198(0): 628-

634.

Bagherzadeh, M., M. Pirmoradian, et al. (2014). "Electrochemical Detection of Pb and

Cu by Using DTPA Functionalized Magnetic Nanoparticles." Electrochimica

Acta 115(0): 573-580.

Bao, H., E. Wang, et al. (2006). "One-Pot Synthesis of CdTe Nanocrystals and Shape

Control of Luminescent CdTe–Cystine Nanocomposites." Small2(4): 476-480.

Bergeron, D. E., O. Coskuner, et al. (2008). "Ligand exchange reactions in the formation

of diphosphine-protected gold clusters." The Journal of Physical Chemistry C

112(33): 12808-12814.

Bertino, M. F., Z.-M. Sun, et al. (2006). "Facile syntheses of monodisperse ultrasmall Au

clusters." The Journal of Physical Chemistry B 110(43): 21416-21418.

Bindhu, M. and M. Umadevi "Green Synthesized Gold Nanoparticles as a Probe for the

Detection of Fe3+ Ions in Water." Journal of Cluster Science: 1-10.

Biver, T., A. Corti, et al. (2012). "Analysis of 4-dimethylaminopyridine (DMAP)-gold

nanoparticles behaviour in solution and of their interaction with calf thymus DNA

and living cells." Journal of Nanoparticle Research 14(2): 1-12.

Blonder, R. (2010). "The story of nanomaterials in modern technology: An advanced

course for chemistry teachers." Journal of Chemical Education 88(1): 49-52.

Bogart, L. K., G. Pourroy, et al. (2014). "Nanoparticles for Imaging, Sensing, and

Therapeutic Intervention." ACS nano 8(4): 3107-3122.

Boisselier, E. and D. Astruc (2009). "Gold nanoparticles in nanomedicine: preparations,

imaging, diagnostics, therapies and toxicity." Chemical Society Reviews 38(6):

1759-1782.

Part II

155

Bolshakova, A., O. Kiselyova, et al. (2001). "Comparative studies of bacteria with an

atomic force microscopy operating in different modes." Ultramicroscopy 86(1):

121-128.

Bouzigues, C., T. Gacoin, et al. (2011). "Biological Applications of Rare-Earth Based

Nanoparticles." ACS Nano 5(11): 8488-8505.

Bruchez, M., M. Moronne, et al. (1998). "Semiconductor nanocrystals as fluorescent

biological labels." Science 281(5385): 2013-2016.

Brust, M., M. Walker, et al. (1994). "Synthesis of thiol-derivatised gold nanoparticles in a

two-phase Liquid-Liquid system." Journal of the Chemical Society, Chemical

Communications(7): 801-802.

Cao, L., J. Sui, et al. (2011). "Generic immunoassay of quinolones: production and

characterization of anti-pefloxacin antibodies as broad selective receptors." Food

Analytical Methods 4(4): 517-524.

Carey, J. S., D. Laffan, et al. (2006). "Analysis of the reactions used for the preparation of

drug candidate molecules." Organic &Biomolecular Chemistry 4(12): 2337-2347.

Carlson, C., S. M. Hussain, et al. (2008). "Unique cellular interaction of silver

nanoparticles: size-dependent generation of reactive oxygen species." The Journal

of Physical Chemistry B 112(43): 13608-13619.

Cassol, C. C., A. P. Umpierre, et al. (2005). "The Role of Pd Nanoparticles in Ionic

Liquid in the Heck Reaction." Journal of the American Chemical Society 127(10):

3298-3299.

Chah, S., M. R. Hammond, et al. (2005). "Gold Nanoparticles as a Colorimetric Sensor

for Protein Conformational Changes." Chemistry & Biology 12(3): 323-328.

Chen, X., Y.-F. Huang, et al. (2008). "Using aptamer–nanoparticle conjugates for cancer

cells detection." Journal of Biomedical Nanotechnology 4(4): 400-409.

Part II

156

Chen, X., Y.-F. Huang, et al. (2008). "Using AptamerNanoparticle Conjugates for Cancer

Cells Detection." Journal of Biomedical Nanotechnology 4(4): 400-409.

Chen, Y. S., Y. C. Hung, et al. (2009). "Assessment of the In Vivo Toxicity of Gold

Nanoparticles." Nanoscale Research Letters 4(8): 858-864.

Cho, J. and F. Caruso (2005). "Investigation of the interactions between ligand-stabilized

gold nanoparticles and polyelectrolyte multilayer films." Chemistry of Materials

17(17): 4547-4553.

Colman, B. P., C. L. Arnaout, et al. (2013). "Low concentrations of silver nanoparticles

in biosolids cause adverse ecosystem responses under realistic field scenario."

PLoS One8(2): e57189.

Cookson, J. (2012). "The preparation of palladium nanoparticles." Platinum Metals

Review 56(2): 83-98.

Corthey, G. n., A. A. Rubert, et al. (2012). "New insights into the chemistry of thiolate-

protected palladium nanoparticles." The Journal of Physical Chemistry C 116(17):

9830-9837.

Coughlin, R. T., C. R. Caldwell, et al. (1981). "A cationic electron spin resonance probe

used to analyze cation interactions with lipopolysaccharide." Biochemical and

Biophysical Research Communications 100(3): 1137-1142.

Coughlin, R. T., S. Tonsager, et al. (1983). "Quantitation of metal cations bound to

membranes and extracted lipopolysaccharide of Escherichia coli." Biochemistry

22(8): 2002-2007.

Craig F. Bohren, D. R. H. (1983). Absorption and scattering of light by small particles.

New York, Wiley: 530.

Croxen, M. A. and B. B. Finlay (2010). "Molecular mechanisms of Escherichia coli

pathogenicity." Nature Reviews Microbiology 8(1): 26-38.

Part II

157

Daniel, M.-C. and D. Astruc (2003). "Gold Nanoparticles: Assembly, Supramolecular

Chemistry, Quantum-Size-Related Properties, and Applications toward Biology,

Catalysis, and Nanotechnology." Chemical Reviews 104(1): 293-346.

Daniel, M. C. and D. Astruc (2004). "Gold Nanoparticles: Assembly, Supramolecular

Chemistry, Quantum-Size-Related Properties, and Applications Toward Biology,

Catalysis, and Nanotechnology." Chemical Reviews 104(1): 293-346.

Dass, A., K. Holt, et al. (2008). "Mass spectrometrically detected statistical aspects of

ligand populations in mixed monolayer Au25L18 nanoparticles." The Journal of

Physical Chemistry C 112(51): 20276-20283.

Davis, R. E., E. Bromels, et al. (1962). "Boron hydrides. III. Hydrolysis of sodium

borohydride in aqueous solution." Journal of the American Chemical Society

84(6): 885-892.

Deng, B., L. Li, et al. (2009). "Pharmacokinetics of pefloxacin mesylate in human urine

using capillary electrophoresis electrochemiluminescence detection." Journal of

Chromatography B 877(24): 2585-2588.

Deng, H.-H., G.-W. Li, et al. (2014). "Thermally treated bare gold nanoparticles for

colorimetric sensing of copper ions." Microchimica Acta 181(9-10): 911-916.

Deng, H.-H., G.-W. Li, et al. (2014). "Thermally treated bare gold nanoparticles for

colorimetric sensing of copper ions." Microchimica Acta: 1-6.

Devani, M., I. T. Patel, et al. (1992). "Spectrophotometric determination of ampicillin

and its dosage forms." Talanta 39(10): 1391-1394.

a -Cruz, M. S., M. a. J. López de Alda, et al. (2003). "Environmental behavior and

analysis of veterinary and human drugs in soils, sediments and sludge." TrAC

Trends in Analytical Chemistry 22(6): 340-351.

Dorofeev, V. (2004). "Infrared spectra and the structure of drugs of the fluoroquinolone

group." Pharmaceutical Chemistry Journal 38(12): 693-697.

Part II

158

Drbohlavova, J., V. Adam, et al. (2009). "Quantum Dots — Characterization, Preparation

and Usage in Biological Systems." International Journal of Molecular Sciences

10(2): 656-673.

Duan, L., Y. Xu, et al. (2008). "Highly sensitive and selective Pd 2+ sensor of

naphthalimide derivative based on complexation with alkynes and thio-

heterocycle." Chemical Communications(47): 6339-6341.

Dupont, J., G. S. Fonseca, et al. (2002). "Transition-metal nanoparticles in imidazolium

ionic liquids: recycable catalysts for biphasic hydrogenation reactions." Journal of

the American Chemical Society 124(16): 4228-4229.

Dupont, J., G. S. Fonseca, et al. (2002). "Transition-Metal Nanoparticles in Imidazolium

Ionic Liquids:  Recycable Catalysts for Biphasic Hydrogenation Reactions."

Journal of the American Chemical Society 124(16): 4228-4229.

El-Kommos, M. E., G. A. Saleh, et al. (2003). "Spectrofluorometric determination of certain

quinolone antibacterials using metal cHeLation." Talanta 60(5): 1033-1050.

El-Safty, S. A. and M. Shenashen (2013). "Optical mesosensor for capturing of Fe (III)

and Hg (II) ions from water and physiological fluids." Sensors and Actuators B:

Chemical 183: 58-70.

El Badawy, A. M., R. G. Silva, et al. (2010). "Surface charge-dependent toxicity of silver

nanoparticles." Environmental Science & Technology 45(1): 283-287.

Fardella, G., P. Barbetti, et al. (1995). "Quantitative analysis of fluoroquinolones by 1 H-and

19 F-NMR spectroscopy." International Journal of Pharmaceutics 121(1): 123-127.

Fayet-Moore, F., P. Petocz, et al. (2014). "Micronutrient Status in Female University

Students: Fe(III), Zinc, Copper, Selenium, Vitamin B12 and Folate." Nutrients

6(11): 5103-5116.

Part II

159

Fisher, J. F., S. O. Meroueh, et al. (2005). "Bacterial Resistance to β-Lactam

Antibiotics:Compelling Opportunism, Compelling Opportunity†." Chemical

Reviews 105(2): 395-424.

Frens, G. (1973). "An experiment concerning the dispersion forces between very small

metal spheres." Physics Letters A 44(3): 208-210.

G. Georgopoulos, A. R., MJ Yonone-Lioy, RE Opiekun, PJ Lioy, P (2001).

"Environmental copper: its dynamics and human exposure issues." Journal of

Toxicology and Environmental Health Part B: Critical Reviews 4(4): 341-394.

Gandubert, V. J. and R. B. Lennox (2005). "Assessment of 4-(dimethylamino) pyridine

as a capping agent for gold nanoparticles." Langmuir 21(14): 6532-6539.

Gao, S., N. Koshizaki, et al. (2009). "Innovative platform for transmission localized

surface plasmon transducers and its application in detecting heavy metal Pd (II)."

Analytical Chemistry 81(18): 7703-7712.

Gao, Y., N. Shao, et al. (2011). "Catalytic activities of subnanometer gold clusters (Au16–

Au18, Au20, and Au27–Au35) for CO oxidation." ACS nano 5(10): 7818-7829.

Garrett, C. E. and K. Prasad (2004). "The art of meeting palladium specifications in

active pharmaceutical ingredients produced by Pd‐catalyzed reactions." Advanced

Synthesis & Catalysis 346(8): 889-900.

Gittins, D. I. and F. Caruso (2001). "Spontaneous Phase Transfer of Nanoparticulate

Metals from Organic to Aqueous Media." Angewandte Chemie International

Edition 40(16): 3001-3004.

Gnanaprakash, G., S. Mahadevan, et al. (2007). "Effect of initial pH and temperature of

Fe(III) salt solutions on formation of magnetite nanoparticles." Materials

Chemistry and Physics 103(1): 168-175.

Part II

160

Grace, A. N. and K. Pandian (2007). "Antibacterial efficacy of aminoglycosidic

antibiotics protected gold nanoparticles—A brief study." Colloids and Surfaces A:

Physicochemical and Engineering Aspects 297(1): 63-70.

Gu, H., P. Ho, et al. (2003). "Presenting vancomycin on nanoparticles to enhance

antimicrobial activities." Nano letters 3(9): 1261-1263.

Hattori, Y., S. Mukasa, et al. (2011). "Synthesis of zinc and zinc oxide nanoparticles from

zinc electrode using plasma in liquid." Materials Letters 65(2): 188-190.

Hong, S., G. Shafai, et al. (2011). "Toward an understanding of ligand selectivity in

nanocluster synthesis." The Journal of Physical Chemistry C 115(30): 14478-

14487.

Hong, Y., Y. M. Huh, et al. (2012). "Nanobiosensors Based on Localized Surface

Plasmon Resonance for Biomarker Detection." Journal of Nanomaterials.

Hua, M., S. J. Zhang, et al. (2012). "Heavy metal removal from water/wastewater by

nanosized metal oxides: A review." Journal of Hazardous Materials 211: 317-331.

Huang, H., K. Wang, et al. (2004). "Design of a Modular‐Based Fluorescent Conjugated

Polymer for Selective Sensing." Angewandte Chemie International Edition

43(42): 5635-5638.

Huang, L.-S. and K.-C. Lin (2001). "Detection of Fe(III) species using inductively

coupled plasma mass spectrometry under cold plasma temperature conditions."

Spectrochimica Acta Part B: Atomic Spectroscopy 56(1): 123-128.

Hudgens, J. W., J. M. Pettibone, et al. (2011). "Reaction mechanism governing formation

of 1, 3-bis (diphenylphosphino) propane-protected gold nanoclusters." Inorganic

Chemistry 50(20): 10178-10189.

Hutchings, G. J., M. Brust, et al. (2008). "Gold-an introductory perspective." Chemical

Society Reviews 37(9): 1759-1765.

Part II

161

Ivask, A., A. ElBadawy, et al. (2013). "Toxicity mechanisms in Escherichia coli vary for

silver nanoparticles and differ from ionic silver." ACS Nano8(1): 374-386.

Iwasawa, T., M. Tokunaga, et al. (2004). "Homogeneous palladium catalyst suppressing

Pd black formation in air oxidation of alcohols." Journal of the American

Chemical Society 126(21): 6554-6555.

Jadzinsky, P. D., G. Calero, et al. (2007). "Structure of a Thiol Monolayer-Protected Gold

Nanoparticle at 1.1 Å Resolution." Science 318(5849): 430-433.

Jamieson, T., R. Bakhshi, et al. (2007). "Biological applications of quantum dots."

Biomaterials 28(31): 4717-4732.

Jang, K.-D., A. Ortega, et al. (2012). "Effect of Lithium Ions on Copper Nanoparticle

Size, Shape, and Distribution." Journal of Nanotechnology 2012.

Jiang, J., H. Jiang, et al. (2011). "A colorimetric and ratiometric fluorescent probe for

palladium." Organic Letters 13(18): 4922-4925.

Ju, J. and W. Chen (2014). "Synthesis of highly fluorescent nitrogen-doped graphene

quantum dots for sensitive, label-free detection of Fe (III) in aqueous media."

Biosensors and Bioelectronics 58: 219-225.

Jung, J. H., J. H. Lee, et al. (2011). "Functionalized magnetic nanoparticles as

chemosensors and adsorbents for toxic metal ions in environmental and biological

fields." Chemical Society Reviews 40(9): 4464-4474.

Kabir, N., H. Ali, et al. (2014). "Silymarin coated gold nanoparticles ameliorates CCl 4-

induced hepatic injury and cirrhosis through down regulation of hepatic stellate

cells and attenuation of Kupffer cells." RSC Advances 4(18): 9012-9020.

Kaplan, O., N. C. Yildirim, et al. (2011). "Assessment of some heavy metals in drinking

water samples of Tunceli, Turkey." Journal of Chemistry 8(1): 276-280.

Part II

162

Kielhorn, J., C. Melber, et al. (2002). "Palladium–a review of exposure and effects to

human health." International Journal of Hygiene and Environmental Health

205(6): 417-432.

Kim, K.-S., D. Demberelnyamba, et al. (2003). "Size-Selective Synthesis of Gold and

Platinum Nanoparticles Using Novel Thiol-Functionalized Ionic Liquids."

Langmuir 20(3): 556-560.

Kim, K. S., S. B. Suh, et al. (2002). "Assembling phenomena of calix [4] hydroquinone

nanotube bundles by one-dimensional short hydrogen bonding and displaced π-π

stacking." Journal of the American Chemical Society 124(47): 14268-14279.

Kim, Y., R. C. Johnson, et al. (2001). "Gold nanoparticle-based sensing of

“spectroscopically silent” heavy metal ions." Nano Letters1(4): 165-167.

Kirubaharan, C. J., D. Kalpana, et al. (2012). "Biomediated Silver Nanoparticles for the

Highly Selective Copper(II) Ion Sensor Applications." Industrial & Engineering

Chemistry Research 51(21): 7441-7446.

Knoppe, S., J. Boudon, et al. (2011). "Size exclusion chromatography for semipreparative

scale separation of Au38 (SR) 24 and Au40 (SR) 24 and larger clusters."

Analytical Chemistry 83(13): 5056-5061.

Lara, F. J., A. M. García-Campaña, et al. (2006). "Multiresidue method for the determination

of quinolone antibiotics in bovine raw milk by capillary electrophoresis-tandem mass

spectrometry." Analytical Cchemistry 78(22): 7665-7673.

Larguinho, M. and P. V. Baptista (2012). "Gold and silver nanoparticles for clinical diagnostics

- From genomics to proteomics." Journal of Proteomics 75(10): 2811-2823.

Lee, H.-C., T.-H. Chen, et al. (2012). "Novel core etching technique on synthesized gold

nanoparticles for colorimetric detection of dopamine biosample". Nano/Micro

Engineered and Molecular Systems (NEMS), 2012 7th IEEE International

Conference on, IEEE.

Part II

163

Leszczynska, E., R. Koncki, et al. (1996). "Biosensors with immobilized penicillin

aminohydrolase and penicillinase for determination of -lactam antibiotics."

Chemia Analityczna 41: 839-844.

Li, J., X. Li, et al. (2003). "Au20: A tetrahedral cluster." Science 299(5608): 864-867.

Link, S. and M. A. El-Sayed (1999). "Spectral properties and relaxation dynamics of

surface plasmon electronic oscillations in gold and silver nanodots and nanorods."

The Journal of Physical Chemistry B 103(40): 8410-8426.

Liu, J.-F., L.-G. Zhang, et al. (1998). "Fabrication of colloidal gold micro-patterns using

photolithographed self-assembled monolayers as templates." Thin Solid Films

327–329(0): 176-179.

Liu, Q., Q. Zhou, et al. (2014). "Nanomaterials for analysis and monitoring of emerging

chemical pollutants." TrAC Trends in Analytical Chemistry 58: 10-22.

Liu, T. Z., S. D. Lee, et al. (1979). "Toxicity of palladium." Toxicology Letters 4(6): 469-

473.

Liu, X., J. G. Worden, et al. (2006). "Kinetic study of gold nanoparticle growth in

solution by Brust-Schiffrin reaction." Journal of Nanoscience and

Nanotechnology 6(4): 1054-1059.

Lo, C.-t. F., K. Karan, et al. (2007). "Kinetic studies of reaction between sodium

borohydride and methanol, water, and their mixtures." Industrial & Engineering

Chemistry Research 46(17): 5478-5484.

Locatelli, C. (2006). "Possible interference in the sequential voltammetric determination

at trace and ultratrace concentration level of platinum group metals (PGMs) and

lead: Application to environmental matrices." Electrochimica Acta 52(2): 614-

622.

Locatelli, C. (2007). "Voltammetric Analysis of Trace Levels of Platinum Group Metals–

Principles and Applications." Electroanalysis 19(21): 2167-2175.

Part II

164

Lu, L. T., L. D. Tung, et al. (2008). "Size and shape control for water-soluble magnetic

cobalt nanoparticles using polymer ligands." Journal of Materials Chemistry

18(21): 2453-2458.

Lu, S., Y. Zhang, et al. (2006). "Preparation of anti-pefloxacin antibody and development

of an indirect competitive enzyme-linked immunosorbent assay for detection of

pefloxacin residue in chicken liver." Journal of Agricultural and Food Chemistry

54(19): 6995-7000.

Magano, J. and J. R. Dunetz (2011). "Large-scale applications of transition metal-

catalyzed couplings for the synthesis of pharmaceuticals." Chemical Reviews

111(3): 2177-2250.

Mah C, Z. I., Fraites TJ, Dobson J, Batich C, Byrne BJ (2000). "Microsphere-mediated

delivery of recombinant AAV vectors in vitro and in vivo." Molecular Therapy

1(5): S239.

Mahtab, R., J. P. Rogers, et al. (1995). "Protein-Sized Quantum-Dot Luminescence Can

Distinguish between Straight, Bent, and Kinked Oligonucleotides." Journal of the

American Chemical Society 117(35): 9099-9100.

Malavolta, M., R. Giacconi, et al. (2010). "Plasma copper/zinc ratio: an

inflammatory/nutritional biomarker as predictor of all-cause mortality in elderly

population." Biogerontology 11(3): 309-319.

Matagne, A., J. Lamotte-Brasseur, et al. (1998). "Catalytic properties of class A beta-

lactamases: efficiency and diversity." Journal of Biochemistry 330(2): 581-598.

Matsukawa, K., S. Watase, et al. (2006). Preparation of metal nano-particles containing

organic-inorganic hybrid thin films and their application.

Mehta, V. N., S. K. Kailasa, et al. (2014). "Sensitive and selective colorimetric sensing of

Fe 3+ ion by using p-amino salicylic acid dithiocarbamate functionalized gold

nanoparticles." New Journal of Chemistry 38(4): 1503-1511.

Part II

165

Meincken, M., D. Holroyd, et al. (2005). "Atomic force microscopy study of the effect of

antimicrobial peptides on the cell envelope of Escherichia coli." Antimicrobial

Agents and Chemotherapy 49(10): 4085-4092.

Miles, D. T. and R. W. Murray (2003). "Temperature-dependent quantized double layer

charging of monolayer-protected gold clusters." Analytical Chemistry 75(6):

1251-1257.

Montay, G., Y. Blain, et al. (1983). "High-performance liquid chromatography of

pefloxacin and its main active metabolites in biological fluids." Journal of

Chromatography B: Biomedical Sciences and Applications 272: 359-365.

Morones, J. R., J. L. Elechiguerra, et al. (2005). "The bactericidal effect of silver

nanoparticles." Nanotechnology 16(10): 2346.

Mostafa, S., M. El-Sadek, et al. (2002). "Spectrophotometric determination of

ciprofloxacin, enrofloxacin and pefloxacin through charge transfer complex

formation." Journal of Pharmaceutical and Biomedical Analysis 27(1): 133-142.

Mulvaney, P. (1996). "Surface Plasmon Spectroscopy of Nanosized Metal Particles."

Langmuir 12(3): 788-800.

Murillo, J. A., J. M. Lemus, et al. (1994). "Spectrofluorimetric analysis of cefoxitin in

pharmaceutical dosage." Talanta 41(4): 557-563.

Murray, C. B., D. J. Norris, et al. (1993). "Synthesis and characterization of nearly

monodisperse CdE (E = S, Se, Te) semiconductor nanocrystallites." Journal of the

American Chemical Society 115(19): 8706-8715.

Nandakumar, R., C. E. Santo, et al. (2011). "Quantitative proteomic profiling of the

Escherichia coli response to metallic copper surfaces." Biometals 24(3): 429-444.

Neely, A., C. Perry, et al. (2009). "Ultrasensitive and highly selective detection of

Al heimer’s disease biomarker using two-photon Rayleigh scattering properties

of gold nanoparticle." ACS Nano 3(9): 2834-2840.

Part II

166

Nikiyan, H., A. Vasilchenko, et al. (2010). "AFM investigations of various disturbing

factors on bacterial cells." Microscopy: Science, Technology, Applications and

Education. Badajoz, Spain: Formatex Research Center: 523-529.

O'Farrell, N., A. Houlton, et al. (2006). "Silicon nanoparticles: Applications in cell

biology and medicine." International Journal of Nanomedicine 1(4): 451-472.

Ohkubo, Y., S. Seino, et al. (2014). "Effect of decrease in the size of Pt nanoparticles

using sodium phosphinate on electrochemically active surface area." Journal of

Nanoparticle Research 16(2): 1-9.

Ohyama, J., Y. Hitomi, et al. (2009). "Size Controlled Synthesis of Gold Nanoparticles

by Porphyrin with Four Sulfur Atoms." Topics in Catalysis 52(6-7): 852-859.

Ooishi, M. and M. Miyao (1997). "Antibiotic sensitivity of recent clinical isolates from

patients with ocular infections." Ophthalmologica 211(Suppl. 1): 15-24.

Patil, R., M. Kokate, et al. (2011). "A novel one step synthesis of silver nanoparticles

using room temperature ionic liquid and their biocidal activity." Comptes Rendus

Chimie 14(12): 1122-1127.

Pérez-López, B. and A. Merkoçi (2011). "Nanomaterials based biosensors for food

analysis applications." Trends in Food Science and Technology 22(11): 625-639.

Perry, C. C., M. Weatherly, et al. (2009). "Atomic force microscopy study of the

antimicrobial activity of aqueous garlic versus ampicillin against Escherichia coli

and Staphylococcus aureus." Journal of the Science of Food and Agriculture

89(6): 958-964.

Pettibone, J. M. and J. W. Hudgens (2010). "Synthetic approach for tunable, size-

selective formation of monodisperse, diphosphine-protected gold nanoclusters."

The Journal of Physical Chemistry Letters 1(17): 2536-2540.

Part II

167

Pettibone, J. M. and J. W. Hudgens (2011). "Gold cluster formation with phosphine

ligands: etching as a size-selective synthetic pathway for small clusters?" ACS

Nano 5(4): 2989-3002.

Pettibone, J. M. and J. W. Hudgens (2012). "Predictive Gold Nanocluster Formation

Controlled by Metal‐Ligand Complexes." Small 8(5): 715-725.

Pettibone, J. M. and J. W. Hudgens (2012). "Reaction network governing diphosphine-

protected gold nanocluster formation from nascent cationic platforms." Physical

Chemistry Chemical Physics 14(12): 4142-4154.

Pettibone, J. M. and N. R. Reardon (2012). "Nucleation products of ligated nanoclusters

unaffected by temperature and reducing agent." Nanoscale 4(18): 5593-5596.

Piao, J., J. Lv, et al. (2014). "A dansyl–rhodamine chemosensor for Fe (III) based on off–

on FRET." Spectrochimica Acta Part A: Molecular and Biomolecular

Spectroscopy 128: 475-480.

Pingali, K. C., S. Deng, et al. (2008). "Effect of Ammonium Nitrate on Nanoparticle Size

Reduction." Research Letters in Nanotechnology 2008.

Pinto, C. G., J. L. P. Pavon, et al. (1995). "Micellar liquid chromatography of zwitterions:

retention mechanism of cephalosporins." Analyst 120(1): 53-62.

Podsiadlo, P., S. Paternel, et al. (2005). "Layer-by-Layer Assembly of Nacre-like

Nanostructured Composites with Antimicrobial Properties." Langmuir 21(25):

11915-11921.

Poole, C. F. (2003). "New trends in solid-phase extraction." TrAC Trends in Analytical

Chemistry 22(6): 362-373.

Prathna, T., N. Chandrasekaran, et al. (2011). "Studies on aggregation behaviour of silver

nanoparticles in aqueous matrices: effect of surface functionalization and matrix

composition." Colloids and Surfaces A: Physicochemical and Engineering

Aspects 390(1): 216-224.

Part II

168

Prichard, H. M. and P. C. Fisher (2012). "Identification of platinum and palladium

particles emitted from vehicles and dispersed into the surface environment."

Environmental Science & Technology 46(6): 3149-3154.

Rademacher, C. and B. Masepohl (2012). "Copper-responsive gene regulation in

bacteria." Microbiology 158(Pt 10): 2451-2464.

Rasouli, S., S. Davaran, et al. (2014). "Positively charged functionalized silica

nanoparticles as nontoxic carriers for triggered anticancer drug release." Designed

Monomers and Polymers 17(3): 227-237.

Ren, W. X., T. Pradhan, et al. (2012). "Rapid responsive palladium sensor under mild

condition." Sensors and Actuators B: Chemical 171: 1277-1282.

Rogach, A. L. (2000). "Nanocrystalline CdTe and CdTe(S) particles: Wet chemical

preparation, size-dependent optical properties and perspectives of optoelectronic

applications." Materials Science and Engineering B: Solid-State Materials for

Advanced Technology 69: 435-440.

Rogach, A. L., A. Eychmüller, et al. (2007). "Infrared-Emitting Colloidal Nanocrystals:

Synthesis, Assembly, Spectroscopy, and Applications." Small 3(4): 536-557.

Rosen, J. E., L. Chan, et al. (2012). "Fe(III) oxide nanoparticles for targeted cancer

imaging and diagnostics." Nanomedicine: Nanotechnology, Biology and

Medicine 8(3): 275-290.

Saha, K., S. S. Agasti, et al. (2012). "Gold nanoparticles in chemical and biological

sensing." Chemical Reviews 112(5): 2739-2779.

Sanderson, H., D. J. Johnson, et al. (2004). "Ranking and prioritization of environmental

risks of pharmaceuticals in surface waters." Regulatory Toxicology and

Pharmacology 39(2): 158-183.

Santoro, M. I. R., N. M. Kassab, et al. (2006). "Quantitative determination of

gatifloxacin, levofloxacin, lomefloxacin and pefloxacin fluoroquinolonic

Part II

169

antibiotics in pharmaceutical preparations by high-performance liquid

chromatography." Journal of Pharmaceutical and Biomedical Analysis 40(1): 179-

184.

Scheinberg, I. H. and I. Sternlieb (1996). "Wilson disease and idiopathic copper

toxicosis." The American Journal of Clinical Nutrition 63(5): 842S-845S.

Scheurell, M., S. Franke, et al. (2009). "Occurrence of diclofenac and its metabolites in

surface water and effluent samples from Karachi, Pakistan." Chemosphere 77(6):

870-876.

Schwarze, T., H. Müller, et al. (2007). "Luminescence Detection of Open‐Shell

Transition‐Metal Ions by Photoinduced Electron Transfer Controlled by Internal

Charge Transfer of a Receptor." Angewandte Chemie International Edition

46(10): 1671-1674.

Selke, S., M. Scheurell, et al. (2010). "Identification and enantioselective gas

chromatographic mass-spectrometric separation of desmethylnaproxen, the main

metabolite of the drug naproxen, as a new environmental contaminant." Journal of

Chromatography A 1217(3): 419-423.

Semetey, V., C. Didierjean, et al. (2002). "Self‐Assembling Organic Nanotubes from

Enantiopure Cyclo‐N, N′‐Linked Oligourease: Design, Synthesis, and Crystal

Structure." Angewandte Chemie 114(11): 1975-1978.

Shah, M. R., S. Ali, et al. (2014). "Morphological analysis of the antimicrobial action of

silver and gold nanoparticles stabilized with ceftriaxone on Escherichia coli using

atomic force microscopy." New Journal of Chemistr y38(11): 5633-5640.

Shamsipur, M., Z. Talebpour, et al. (2002). "Monitoring of ampicillin and its related

substances by NMR." Journal of Pharmaceutical and Biomedical Analysis 30(4):

1075-1085.

Part II

170

Sharma, A., R. Khosla, et al. (1994). "Fluoroquinolones: antimicrobial agents of the

90’s." Indian Journal of Pharmacology 26(4): 249.

Sharma, V. K., R. A. Yngard, et al. (2009). "Silver nanoparticles: green synthesis and

their antimicrobial activities." Advances in Colloid and Interface Science 145(1):

83-96.

Shinkai, M., M. Yanase, et al. (1999). "Intracellular hyperthermia for cancer using

magnetite cationic liposomes." Journal of Magnetism and Magnetic Materials

194(1-3): 176-184.

Shiraishi, Y., M. Ikeda, et al. (2011). "One-pot synthesis of imines from alcohols and

amines with TiO2 loading Pt nanoparticles under UV irradiation." Chemical

Communications 47(16): 4811-4813.

Siripinyanond, A., S. Worapanyanond, et al. (2005). "Field-flow fractionation-inductively

coupled plasma mass spectrometry: an alternative approach to investigate metal-

humic substances interaction." Environmental Science & Technology 39(9):

3295-3301.

Sondi, I. and B. Salopek-Sondi (2004). "Silver nanoparticles as antimicrobial agent: a

case study on E. coli as a model for Gram-negative bacteria." Journal of Colloid

and Interface Science 275(1): 177-182.

Song, F., A. L. Garner, et al. (2007). "A highly sensitive fluorescent sensor for palladium

based on the allylic oxidative insertion mechanism." Journal of the American

Chemical Society 129(41): 12354-12355.

Sun, S., C. B. Murray, et al. (2000). "Monodisperse FePt Nanoparticles and

Ferromagnetic FePt Nanocrystal Superlattices." Science 287(5460): 1989-1992.

Sun, S. and H. Zeng (2002). "Size-controlled synthesis of magnetite nanoparticles."

Journal of the American Chemical Society 124(28): 8204-8205.

Part II

171

Sun, X., L. Wu, et al. (2013). "Longitudinal surface plasmon resonance assay enhanced

by magnetosomes for simultaneous detection of Pefloxacin and Microcystin-LR

in seafoods." Biosensors and Bioelectronics 47: 318-323.

Sun, Y. F., S. B. Liu, et al. (2012). "Metal Oxide Nanostructures and Their Gas Sensing

Properties: A Review." Sensors 12(3): 2610-2631.

Swoboda, S., K. Oberdorfer, et al. (2003). "Tissue and serum concentrations of

levofloxacin 500 mg administered intravenously or orally for antibiotic

prophylaxis in biliary surgery." Journal of Antimicrobial Chemotherapy 51(2):

459-462.

Taglietti, A., Y. A. Diaz Fernandez, et al. (2012). "Antibacterial activity of glutathione-

coated silver nanoparticles against gram positive and gram negative bacteria."

Langmuir 28(21): 8140-8148.

Tang, B., H. Zhang, et al. (2004). "Determination of trace palladium with salicyladehyde

furfuralhydrazone (SAFH) by catalytic kinetic spectrofluorimetry." Analytical

Letters 37(6): 1219-1231.

Thomson, A. M., J. T. Rogers, et al. (1999). "Fe(III)-regulatory proteins, Fe(III)-

responsive elements and ferritin mRNA translation." The International Journal of

Biochemistry & Cell Biology 31(10): 1139-1152.

Toikkanen, O., S. Carlsson, et al. (2009). "Solvent-Dependent Stability of Monolayer-

Protected Au38 Clusters." The Journal of Physical Chemistry Letters 1(1): 32-37.

Tourneux, E., H. Gornitzka, et al. (2007). "Palladate Salts from Chiral Pyridinium Ionic

Liquids: Synthesis and Crystal Structures." Molecules 12(8): 1940-1949.

Tripathy, S. K., J. Y. Woo, et al. (2013). "Colorimetric detection of Fe (III) ions using

label-free gold nanoparticles and acidic thiourea mixture." Sensors and Actuators

B: Chemical 181: 114-118.

Part II

172

Tshikhudo, T. R., D. Demuru, et al. (2005). "Molecular recognition by calix[4]arene-

modified gold nanoparticles in aqueous solution." Angewandte Chemie -

International Edition 44(19): 2913-2916.

Turkevich, J., P. C. Stevenson, et al. (1951). "A study of the nucleation and growth

processes in the synthesis of colloidal gold." Discussions of the Faraday Society

11(0): 55-75.

Tyczkowska, K. L., R. D. Voyksner, et al. (1992). "Solvent degradation of cloxacillin in

vitro: Tentative identification of degradation products using thermospray liquid

chromatography-mass spectrometry." Journal of Chromatography A594(1–2):

195-201.

Uauy, R., M. Olivares, et al. (1998). "Essentiality of copper in humans." The American

Journal of Clinical Nutrition 67(5): 952S-959S.

Umbreit, J. (2005). "Fe(III) deficiency: a concise review." American Journal of

Hematology 78(3): 225-231.

Ung, D., L. D. Tung, et al. (2009). "Variant shape growth of nanoparticles of metallic Fe-

Pt, Fe-Pd and Fe-Pt-Pd alloys." CrystEngComm 11(7): 1309-1316.

Valodkar, M., S. Modi, et al. (2011). "Synthesis and anti-bacterial activity of Cu, Ag and

Cu–Ag alloy nanoparticles: A green approach." Materials Research Bulletin

46(3): 384-389.

Veiopoulou, C., P. Ioannou, et al. (1997). "Application of terbium sensitized fluorescence

for the determination of fluoroquinolone antibiotics pefloxacin, ciprofloxacin and

norfloxacin in serum." Journal of Pharmaceutical and Biomedical Analysis15(12):

1839-1844.

Verma, P. K., A. Giri, et al. (2010). "Superparamagnetic fluorescent nickel-enzyme

nanobioconjugates: synthesis and characterization of a novel multifunctional

biological probe." Journal of Materials Chemistry 20(18): 3722-3728.

Part II

173

Vertelov, G., Y. A. Krutyakov, et al. (2008). "A versatile synthesis of highly bactericidal

Myramistin® stabilized silver nanoparticles." Nanotechnology 19(35): 355707.

Vigderman, L. and E. R. Zubarev (2013). "Therapeutic platforms based on gold

nanoparticles and their covalent conjugates with drug molecules." Advanced Drug

Delivery Reviews 65(5): 663-676.

Wang, B., J. Hai, et al. (2010). "Selective Detection of Fe(III) (III) by

Rhodamine‐Modified Fe3O4 Nanoparticles." Angewandte Chemie International

Edition 49(27): 4576-4579.

Wang, J. and K. Varughese (1987). "Determination of traces of palladium by adsorptive

stripping voltammetry of the dimethylglyoxime complex." Analytica Chimica

Acta 199: 185-189.

Wang, R., F. Yu, et al. (2012). "A turn-on fluorescent probe based on hydroxylamine

oxidation for detecting ferric ion selectively in living cells." Chemical

Communications 48(43): 5310-5312.

Wang, S., S. Sato, et al. (2003). "Preparation of hexagonal-close-packed colloidal crystals

of hydrophilic monodisperse gold nanoparticles in bulk aqueous solution."

Chemistry of Materials 15(12): 2445-2448.

Wang, Y. and H. Yang (2005). "Synthesis of CoPt Nanorods in Ionic Liquids." Journal of

the American Chemical Society 127(15): 5316-5317.

Weissleder, R., G. Elizondo, et al. (1990). "Ultrasmall superparamagnetic Fe(III) oxide:

Characterization of a new class of contrast agents for MR imaging." Radiology

175(2): 489-493.

Wilson, B. A., V. H. Smith, et al. (2003). "Effects of three pharmaceutical and personal

care products on natural freshwater algal assemblages." Environmental Science &

Technology 37(9): 1713-1719.

Part II

174

Wishart, D. (1984). "Recent advances in antimicrobial drugs: the penicillins." Journal of

the American Veterinary Medical Association 185(10): 1106-1108.

Wu, P., Y. Li, et al. (2009). "CdTe Quantum Dots (QDs) Based Kinetic Discrimination of

Fe2+ and Fe3+, and CdTe QDs-Fenton Hybrid System for Sensitive

Photoluminescent Detection of Fe2+." Analytical Chemistry 81(15): 6252-6257.

Wu, S.-P., Y.-P. Chen, et al. (2011). "Colorimetric detection of Fe 3+ ions using

pyrophosphate functionalized gold nanoparticles." Analyst 136(9): 1887-1891.

Xiang, G., Y. Zhang, et al. (2010). "Determination of trace copper in food samples by

flame atomic absorption spectrometry after solid phase extraction on modified

soybean hull." Journal of Hazardous Materials 179(1): 521-525.

Xiao, Q., H. Gao, et al. (2012). "Gold nanoparticle-based optical probes for sensing

aminothiols." TrAC - Trends in Analytical Chemistry 40: 64-76.

Xie, Y., Y. Song, et al. (2010). "Near-infrared spectroscopy quantitative determination of

Pefloxacin mesylate concentration in pharmaceuticals by using partial least

squares and principal component regression multivariate calibration."

Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy75(5):

1535-1539.

Yang, J., G. Zhou, et al. (1998). "Study on the fluorescence characteristics of alkaline

degradation of cefadroxil, cephradine, cefotaximum sodium and amoxicillini."

Analytical Letters 31(6): 1047-1060.

Yang, X., S. Zhu, et al. (2014). "Novel and remarkable enhanced-fluorescence system

based on gold nanoclusters for detection of tetracycline." Talanta 122: 36-42.

Yang, Z., X. Wang, et al. (2008). "Capillary electrophoresis–chemiluminescence

determination of norfloxacin and prulifloxacin." Analytica Chimica Acta 623(2):

231-237.

Part II

175

Yantasee, W., K. Hongsirikarn, et al. (2008). "Direct detection of Pb in urine and Cd, Pb,

Cu, and Ag in natural waters using electrochemical sensors immobilized with

DMSA functionalized magnetic nanoparticles." Analyst 133(3): 348-355.

Zhang, H., X. Wang, et al. (2000). "Functional crystals: Search criteria and design

principles." Journal of Solid State Chemistry 152(1): 191-198.

Zhang, Y. L., R. W. Cattrall, et al. (2011). "Gold, an alternative to platinum group metals

in automobile catalytic converters." Gold Bulletin 44(3): 145-153.

Zhao, Z. and Z. Gao (1988). "Single sweep polarography of palladium-dimethylglyoxime

complex." Journal of Electroanalytical Chemistry and Interfacial Electrochemistry

256(1): 65-75.

Zhou, Y., J. Zhang, et al. (2012). "A highly sensitive and selective “off–on” chemosensor

for the visual detection of Pd 2+ in aqueous media." Sensors and Actuators B:

Chemical 171: 508-514.

Zhu, Y., S. H. A. Jang, et al. (2011). "An Efficient and Recyclable Catalytic System

Comprising Nano-Iridium(0) and a Pyridinium Salt of nido-Carboranyldiph

osphine for the Synthesis of One-Dimensional Boronate Esters via Hydroboration

Reaction." Organometallics31 (7): 2589-2596.

Conclusions

176

CONCLUSIONS

We have shown that the size selectivity of diphosphine capped gold clusters is dependent

on size and flexibility of spacer groups. Results revealed that the smallest spacer

(bis(diphenylphosphino)propane/L3) exhibits the greatest size selectivity towards cluster

formation of smaller core sizes in all solvents used as compared to other flexible and

rigid spacer. The molecular recognition of 3 synthesized metal nanoparticles was also

developed.We have successfully established a quick and sensitive optical sensor for the

selective detection of Cu(I), based on Para-formylpyridinium propylthioacetate ligand

stabilized silver nanoparticles. The applied protocol is feasible for detection of copper in

both the T-lymphocytes and human blood plasma. This nanosensor offers fast, simple and

cost efficient method for selective detection of Cu(I) as well as 6-APA. Furthermore, a

robust method for the synthesis of 1-(3-(acetylthio)propyl)pyrazin-1-ium protected stable

AuNPs was developed. The novel AuNPs were applied for selectively sensing of Fe(III)

on the basis of aggregation-induced absorbance quenching, and colorimetric detection of

Pd(II). Finally, weoptimized a facile and rapid nanosensor based on 1-(3-

(acetylthio)propyl)-4-(dimethylamino)pyridin-1-ium capped AuNPs for the detection of

Pefloxacin mesylate. The optical response of Pefloxacin mesylate is distinctive, and

follows linear correlation in a wide range of the drug with comparable LOD as compared

to other reported methods. Importantly, these detection systems worked precisely with

good selectivity towards analytes in the presence of many interfering species, as well as

in tap water for drinking, and in physiologically important human blood plasma samples.

A series of novel dihydropyridine derivatives was synthesized, and it was screened by

cytotoxicity, antioxidant and urease inhibition assays. Two compounds were found highly

potent as antioxidant, while one significantly cytotoxic. Moreover, to specifically target

Acanthamoeba, we achieved the synthesis of photosensitising compound porphyrin and

conjugated it with mannose and tested its photochemotherapeutic efficacy in vitro. The

results revealed that mannose-conjugated porphyrin produced potent trophicidal effects

and blocked excystation of the microbe. These data suggest that mannose-conjugated

porphyrin has application for the targeted photodynamic therapy of Acanthamoeba

infections. The finding of this study will help in the development of new chemosensors

based on nanoparticles and also new antibacterial drug development.

List of Publications

177

LIST OF PUBLICATIONS

1) Aqeel, Y., R. Siddiqui, A. Anwar, et al. (2015). "Photochemotherapeutic strategy

against Acanthamoeba infections." Antimicrobial Agents and Chemotherapy:

AAC. 05126-05114.

2) Anwar, A., A. Hameed, et al. (2014). "1, 1-Diphenyl-2-picrylhydrazyl radical

scavenging activity of novel dihydropyridine derivatives." European Journal of

Chemistry 5 (1): 189-191.

3) Hameed, A., A. Anwar, et al. (2013). "Urease inhibition and anticancer activity of

novel polyfunctional 5, 6-dihydropyridine derivatives and their structure-activity

relationship." European Journal of Chemistry 4 (1): 49-52.

4) Hameed, A., A. Anwar, et al. (2012). "Tetra-n-butylammonium fluoride-mediated

dimerization of (α-methylbenzylidene) malononitriles to form polyfunctional 5, 6-

dihydropyridines derivatives under solvent-free conditions." European Journal of

Chemistry 3 (2): 179-185.

5) Anwar, A. M. Shah, et al. (2015). “Optical sensing of Cu(I) in real samples and T-

lymphocytes with Thia-pyridinium capped silver nanoparticles” (Submitted)

6) Anwar, A. M. Shah, et al. (2015). “Atomic Force Microscope based intra-

microbial sensing of Cu(I) by Silver nanoparticles with enhanced antimicrobial

activity” (Submitted)

7) Aqeel, Y., R. Siddiqui, A. Anwar, et al. (2015).” Gold nanoparticle conjugation

enhanced anti-Acanthamoebic effects of chlorhexidine” (Submitted)