1
50 25 12.5 6.25 300 0 200 400 600 800 Parkin W403A Auto-Ub 20min Parkin W403A (nM) % Signal:Background (rS-rB/rB)x100% Development of a Real-Time Homogeneous TR-FRET Ubiquitin Conjugation and Deconjugation Assay Platform By South Bay Bio, 5941 Optical Court, Suite 229, San Jose CA, 95138 Contact [email protected], Tel. (415) 935-3226 Ubiquitin, a highly conserved 76 amino acid protein, is an important post- translational modifier responsible for regulating a wide variety of cellular functions including but not limited to protein degradation and recycling, cell cycle control, and DNA damage repair. Ubiquitination, the modification of proteins by the attachment of ubiquitin or polyubiquitin chains, is catalyzed by a three step process in which ubiquitin is activated by an ATP-dependent E1 activating enzyme, transferred to an E2 conjugating enzyme, then finally conjugated to a target substrate by an E3 ubiquitin ligase. The process is reversed by ubiquitin deconjugating enzymes (DUBs.) Thus far, assay development within the ubiquitin field has suffered a lack of tools for probing the activity of the conjugation pathway. Herein, we report the development of a novel assay platform capable of measuring ubiquitin conjugation and deconjugation in real time in a homogenous, single step assay, utilizing TR-FRET technology. TR-FRET (Time Resolved Fluorescence Resonance Energy Transfer) uses the extended fluorescence emission decay lifetimes typical of rare-earth lanthanides to impart a short time-delay between FRET donor excitation and emission. This delay provides a ŵeaŶs to sepaƌate tƌue sigŶal fƌoŵ shoƌt-lived background fluorescence and reduce interference from compound fluorescence and other assay artifacts. Using ubiquitin labeled with either Europium-Cryptate (donor) or Cyanine5 (acceptor), we demonstrate ubiquitin conjugation and deconjugation measured homogenously in real time, with assaLJs ĐoŵŵoŶlLJ edžhiďitiŶg Z’ ≥ 0.8. We shoǁ edžaŵples of auto-ubiquitination kinetics of several human recombinant E3 ligases of significant interest: MDM2, Parkin, ITCH, NEDD4, and XIAP. Additionally, we are able to show the reversal of this process ďLJ deuďiƋuitiŶatiŶg uďiƋuitiŶated MDM2 ǁith the DUB U“P2, aŶd Đoŵpaƌe U“P2’s kinetics with this substrate versus Ubiquitin-Rhodamine 110. Activity of All Types of E3 Ubiquitin Ligases can be Measured in a Real-Time Homogeneous TR-FRET Assay TR-FRET MDM2 Autoubiquitination: Serial dilutions of MDM2 from 200nM to 12.5nM mixed with UBA1, UBE2D3, trf-Ub mix. Reaction was initiated with addition of Mg-ATP, excited at 304 nm, and emission detected at 620 nm, and 665 nm. Endpoint E3 Ubiquitin Ligase Autoubiquitination: Conjugation reactions can also be measured in an endpoint configuration. Optimal incubation time has been selected from real-time experiments where the rate of conjugation is linear. Setup conditions were similar to a real-time reactions, except 10mM DTT and 2mM EDTA were added to quench the reactions before measurement. Deconjugation Deconjugating enzymes (DUBs) catalyze the removal of ubiquitin from substrate proteins. This reversal of the conjugation cascade affects critical events in the cell proteome such as protein degradation through the proteasome. To date, more than 100 DUBs have been annotated, and many studies have shown their multiple functions in human disease. Current DUB screening reagents largely consist of c-terminal derivatives such as ubiquitin rhodamine 110 and AMC. However, a growing need for more physiologically relevant substrates is emerging as we discover more DUBs with preferential lysine linkage specificities or multimeric ubiquitin chain requirements TR-FRET Tech Rare earth cryptates are macrocyclical structures encasing a rare earth lanthanide atoŵ. LaŶthaŶide ioŶs doŶ’t edžhiďit suitaďle fluoƌesĐeŶĐe pƌopeƌties oŶ theiƌ oǁŶ, aŶd ƌeƋuiƌe iŶĐoƌpoƌatioŶ ǁith oƌgaŶiĐ ŵoieties fuŶĐtioŶiŶg as light-haƌǀestiŶg antennas to collect and transfer energy by intramolecular non-radiative processes. Our cryptate is composed of a macrocycle in a cage-shaped assembly composed of three bipyridine arms, which complex a Eu 3+ ion (Eu 3+ TBP) as shown in the structure below. Unlike other common flavors of rare earth complexes like chelates, cryptates are extremely robust and stable structures that show no sensitivity to photobleaching. This is largely due to cryptates having no dissociation between the complexed ion and the macrocycle, contrary to chelates, which often exhibit uncoordinated bonds with solvent. These properties allow cryptates to perform in stringent conditions, e.g. in presence of strong chelators, of divalent cations (e.g. Mn 2+ , Mg 2+ ), extremes of pH, organic solvents, and high temperatures (PCR or other thermoregulated processes). Introduction TR-FRET MDM2-Ub n Chains Titrated with USP2 CD : 50nM MDM2 was conjugated with trf-Ub chains using UBA1, UBE2D3, and Mg-ATP for 3 hours. TR-FRET MDM2-Ub n chains were then digested using titrating amounts of USP2 CD , from 93nM to 11nM. Initial velocities (delta (sec -1 )) are shown plotted as a function of USP2 CD concentration. Increasing the DUB concentration increases the reaction rate linearly. TR-FRET MDM2-Ub n Chains & USP2 CD Ub-Aldehyde Dose- Response: 50nM MDM2 pre-conjugated with trf-Ub chains was digested with 250nM USP2 CD along with serial dilutions of Ub-Aldehyde. Initial rates of each inhibitor concentration were calculated and normalized relative to the no-inhibition control. Normalized activity was plotted against the log of inhibitor concentration, and fit with a 4 th parameter agonist curve yielding an IC 50 of 152nM. A similar dose-response using 500nM Ubiquitin-Rhodamine 110 substituted for trf- MDM2-Ub n yields similar results. E3 Ligase Autoubiquitination Measured in an Endpoint TR-FRET Assay TR-FRET MDM2-Ub n Digests with USP2 CD & Measures K i Values RING HECT RBR Summary TR-FRET XIAP Autoubiquitination: Serial dilutions of XIAP from 350nM to 43.75nM mixed with UBA1, UBE2D2, trf-Ub mix. Reaction was initiated with addition of Mg-ATP, excited at 304 nm, and emission detected at 620 nm, and 665 nm. TR-FRET ITCH Autoubiquitination: Serial dilutions of ITCH from 130nM to 8.125nM mixed with UBA1, UBE2L3, trf-Ub mix. Reaction was initiated with addition of Mg-ATP, excited at 304 nm, and emission detected at 620 nm, and 665 nm. TR-FRET NEDD4 Autoubiquitination: Serial dilutions of NEDD4 from 50nM to 3.125nM mixed with UBA1, UBE2L3, trf-Ub mix. Reaction was initiated with addition of Mg-ATP, excited at 304 nm, and emission detected at 620 nm, and 665 nm. TR-FRET Parkin W403A Autoubiquitination: Serial dilutions of Parkin W403A from 50nM to 6.25nM mixed with UBA1, UBE2D3, trf-Ub mix. Reaction was initiated with addition of Mg-ATP, excited at 304 nm, and emission detected at 620 nm, and 665 nm. 300nM wt-Parkin shown for comparison. In this report, we show the capabilities of a novel TR-FRET based platform for probing the activities of E3 ubiquitin ligases and deubiquitinating enzymes in real-time and endpoint systems. This technology fills a longstanding gap in the ability of researchers to conduct drug discovery with simple assays tailored for the ubiquitin proteasome system. Unlike existing technologies, our platform does not rely on secondary detection methods such as antibodies. We deŵoŶstƌate this platfoƌŵ’s ƌoďustŶess ǁith assaLJs ĐoŵŵoŶlLJ edžhiďitiŶg Z’ ≥0.8, aŶd shoǁ its adaptability to miniaturized formats, making it ideal for high-throughput screening experiments. We further provide a proof-of-concept for the sLJsteŵ’s utilitLJ aŶd siŵpliĐitLJ iŶ ŵakiŶg K i determinations, and demonstrate that the platform holds its weight against a common DUB substrate, Ubiquitin Rhodamine 110. 0 20 40 60 80 100 -100 -50 0 Ubiquitinated MDM2 vs. USP2 CD Titration USP2 CD (nM) Rate (delta)(sec -1 ) References 0 15 30 45 60 75 90 105 120 135 150 0 1000 2000 3000 MDM2 Autoubiquitination time (min) % Signal:Background (rS-rB/rB)x100% 200nM 100nM 50nM 25nM 12.5nM 15 30 45 60 75 90 105 120 135 150 0.0 0.5 1.0 MDM2 Autoubiquitination Z' time (min) Estimated Z-factor 200nM 100nM 50nM 25nM 12.5nM 0 15 30 45 60 75 90 105 120 135 150 0 200 400 600 800 1000 XIAP Autoubiquitination time (min) % Signal:Background (rS-rB/rB)x100% 350nM 175nM 87.5nM 43.75nM 15 30 45 60 75 90 105 120 135 150 0.0 0.5 1.0 XIAP Autoubiquitination Z' time (min) Estimated Z-factor 350nM 175nM 87.5nM 43.75nM 0 15 30 45 60 75 90 105 120 135 150 0 500 1000 ITCH Autoubiquitination time (min) % Signal:Background (rS-rB/rB)x100% 130nM 65nM 32.5nM 16.25nM 8.125nM 15 30 45 60 75 90 105 120 135 150 0.0 0.5 1.0 ITCH Autoubiquitination Z' time (min) Estimated Z-factor 130nM 65nM 32.5nM 16.25nM 8.125nM 0 15 30 45 60 75 90 105 120 135 150 0 500 1000 1500 Parkin W403A Autoubiquitination time (min) % Signal:Background (rS-rB/rB)x100% 50 nM 25 nM 12.5 nM 6.25 nM 300 nM wt Parkin 15 30 45 60 75 90 105 120 135 150 0.0 0.5 1.0 Parkin W403A Autoubiquitination Z' time (min) Estimated Z-factor 50 nM 25 nM 12.5 nM 6.25 nM 300 nM wt Parkin 0 15 30 45 60 75 90 105 120 135 150 165 180 0 1000 2000 3000 NEDD4 Autoubiquitination time (min) % Signal:Background (rS-rB/rB)x100% 50 nM 25 nM 12.5 nM 6.25 nM 3.125 nM 15 30 45 60 75 90 105 120 135 150 0.0 0.5 1.0 NEDD4 Autoubiquitination Z' time (min) Estimated Z-factor 50 nM 25 nM 12.5 nM 6.25 nM 3.125 nM 200 100 50 25 12.5 0 500 1000 1500 2000 2500 Nedd4 Auto-Ub 30 min NEDD4 (nM) % Signal:Background (rS-rB/rB)x100% 200 100 50 25 12.5 0 500 1000 1500 2000 MDM2 Auto-Ub 30min MDM2 (nM) % Signal:Background (rS-rB/rB)x100% wt Parkin 130 65 32.5 16.25 8.125 0 200 400 600 800 1000 ITCH Auto-Ub 30min ITCH (nM) % Signal:Background (rS-rB/rB)x100% 350 175 87.5 43.75 0 200 400 600 800 XIAP Auto-Ub 15 min XIAP (nM) % Signal:Background (rS-rB/rB)x100% 0.001 0.01 0.1 1 10 0.0 0.5 1.0 Ub-Aldehyde Titration & 250nM USP2 CD Ubiquitin-Aldehyde ( M) Normalized USP2 cd Activity IC 50 = 0.152 M MDM2-UB n UB-RH 110 IC 50 = 0.175 M Amerik, A. Y., & Hochstrasser, M. (2004). Mechanism and function of deubiquitinating enzymes. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research, 1695(1), 189-207. Koyano, F., Okatsu, K., Kosako, H., Tamura, Y., Go, E., Kimura, M., ... & Endo, T. (2014). Ubiquitin is phosphorylated by PINK1 to activate parkin. Nature, 510(7503), 162. Magennis, S. W., Parsons, S., Pikramenou, Z., Corval, A., & Woollins, J. D. (1999). Imidodiphosphinate ligands as antenna units in luminescent lanthanide complexes. Chemical communications, (1), 61-62. Zheng, N., & Shabek, N. (2017). Ubiquitin Ligases: Structure, Function, and Regulation. Annual Review of Biochemistry, (0). Powered by TCPDF (www.tcpdf.org) Powered by TCPDF (www.tcpdf.org) Powered by TCPDF (www.tcpdf.org) Powered by TCPDF (www.tcpdf.org)

Development of a Real-Time Homogeneous TR-FRET Ubiquitin

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Development of a Real-Time Homogeneous TR-FRET Ubiquitin

5 0 2 5 1 2 .5 6 .2 5 3 0 0

0

2 0 0

4 0 0

6 0 0

8 0 0

P a r k inW 4 0 3 A

A u t o -U b 2 0 m in

P a r k inW 4 0 3 A

(n M )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

Development of a Real-Time Homogeneous TR-FRET Ubiquitin

Conjugation and Deconjugation Assay PlatformBy South Bay Bio, 5941 Optical Court, Suite 229, San Jose CA, 95138

Contact [email protected], Tel. (415) 935-3226

Ubiquitin, a highly conserved 76 amino acid protein, is an important post-

translational modifier responsible for regulating a wide variety of cellular functions

including but not limited to protein degradation and recycling, cell cycle control, and

DNA damage repair. Ubiquitination, the modification of proteins by the attachment of

ubiquitin or polyubiquitin chains, is catalyzed by a three step process in which ubiquitin

is activated by an ATP-dependent E1 activating enzyme, transferred to an E2

conjugating enzyme, then finally conjugated to a target substrate by an E3 ubiquitin

ligase. The process is reversed by ubiquitin deconjugating enzymes (DUBs.) Thus far,

assay development within the ubiquitin field has suffered a lack of tools for probing the

activity of the conjugation pathway. Herein, we report the development of a novel

assay platform capable of measuring ubiquitin conjugation and deconjugation in real

time in a homogenous, single step assay, utilizing TR-FRET technology.

TR-FRET (Time Resolved Fluorescence Resonance Energy Transfer) uses the extended

fluorescence emission decay lifetimes typical of rare-earth lanthanides to impart a

short time-delay between FRET donor excitation and emission. This delay provides a

ea s to sepa ate t ue sig al f o sho t-lived background fluorescence and reduce

interference from compound fluorescence and other assay artifacts. Using ubiquitin

labeled with either Europium-Cryptate (donor) or Cyanine5 (acceptor), we demonstrate

ubiquitin conjugation and deconjugation measured homogenously in real time, with

assa s o o l e hi iti g Z’ ≥ 0.8. We sho e a ples of auto-ubiquitination

kinetics of several human recombinant E3 ligases of significant interest: MDM2, Parkin,

ITCH, NEDD4, and XIAP. Additionally, we are able to show the reversal of this process

deu i uiti ati g u i uiti ated MDM2 ith the DUB U“P2, a d o pa e U“P2’s kinetics with this substrate versus Ubiquitin-Rhodamine 110.

Activity of All Types of E3 Ubiquitin Ligases can be Measured in a Real-Time Homogeneous TR-FRET Assay

TR-FRET MDM2 Autoubiquitination: Serial dilutions

of MDM2 from 200nM to 12.5nM mixed with UBA1,

UBE2D3, trf-Ub mix. Reaction was initiated with

addition of Mg-ATP, excited at 304 nm, and emission

detected at 620 nm, and 665 nm.

Endpoint E3 Ubiquitin Ligase Autoubiquitination: Conjugation reactions can also be measured in an endpoint configuration. Optimal incubation time has been selected from real-time experiments

where the rate of conjugation is linear. Setup conditions were similar to a real-time reactions, except 10mM DTT and 2mM EDTA were added to quench the reactions before measurement.

DeconjugationDeconjugating enzymes (DUBs) catalyze the removal of ubiquitin from substrate

proteins. This reversal of the conjugation cascade affects critical events in the cell

proteome such as protein degradation through the proteasome. To date, more than

100 DUBs have been annotated, and many studies have shown their multiple functions

in human disease. Current DUB screening reagents largely consist of c-terminal

derivatives such as ubiquitin rhodamine 110 and AMC. However, a growing need for

more physiologically relevant substrates is emerging as we discover more DUBs with

preferential lysine linkage specificities or multimeric ubiquitin chain requirements

TR-FRET TechRare earth cryptates are macrocyclical structures encasing a rare earth lanthanide

ato . La tha ide io s do ’t e hi it suita le fluo es e e p ope ties o thei o , a d e ui e i o po atio ith o ga i oieties fu tio i g as light-ha esti g antennas to collect and transfer energy by intramolecular non-radiative processes.

Our cryptate is composed of a macrocycle in a cage-shaped assembly composed of

three bipyridine arms, which complex a Eu3+ ion (Eu3+TBP) as shown in the structure

below. Unlike other common flavors of rare earth complexes like chelates, cryptates

are extremely robust and stable structures that show no sensitivity to

photobleaching. This is largely due to cryptates having no dissociation between the

complexed ion and the macrocycle, contrary to chelates, which often exhibit

uncoordinated bonds with solvent. These properties allow cryptates to perform in

stringent conditions, e.g. in presence of strong chelators, of divalent cations (e.g.

Mn2+, Mg2+), extremes of pH, organic solvents, and high

temperatures (PCR or other thermoregulated

processes).

Introduction

TR-FRET MDM2-Ubn Chains Titrated with USP2CD: 50nM

MDM2 was conjugated with trf-Ub chains using UBA1,

UBE2D3, and Mg-ATP for 3 hours. TR-FRET MDM2-Ubn

chains were then digested using titrating amounts of USP2CD,

from 93nM to 11nM. Initial velocities (delta (sec-1)) are

shown plotted as a function of USP2CD concentration.

Increasing the DUB concentration increases the reaction rate

linearly.

TR-FRET MDM2-Ubn Chains & USP2CD Ub-Aldehyde Dose-

Response: 50nM MDM2 pre-conjugated with trf-Ub chains

was digested with 250nM USP2CD along with serial dilutions

of Ub-Aldehyde. Initial rates of each inhibitor concentration

were calculated and normalized relative to the no-inhibition

control. Normalized activity was plotted against the log of

inhibitor concentration, and fit with a 4th parameter agonist

curve yielding an IC50 of 152nM. A similar dose-response

using 500nM Ubiquitin-Rhodamine 110 substituted for trf-

MDM2-Ubn yields similar results.

E3 Ligase Autoubiquitination Measured in an Endpoint TR-FRET Assay

TR-FRET MDM2-Ubn Digests with USP2CD & Measures Ki Values

RING HECT RBR

Summary

TR-FRET XIAP Autoubiquitination: Serial dilutions of

XIAP from 350nM to 43.75nM mixed with UBA1,

UBE2D2, trf-Ub mix. Reaction was initiated with

addition of Mg-ATP, excited at 304 nm, and emission

detected at 620 nm, and 665 nm.

TR-FRET ITCH Autoubiquitination: Serial dilutions of

ITCH from 130nM to 8.125nM mixed with UBA1,

UBE2L3, trf-Ub mix. Reaction was initiated with

addition of Mg-ATP, excited at 304 nm, and emission

detected at 620 nm, and 665 nm.

TR-FRET NEDD4 Autoubiquitination: Serial dilutions

of NEDD4 from 50nM to 3.125nM mixed with UBA1,

UBE2L3, trf-Ub mix. Reaction was initiated with

addition of Mg-ATP, excited at 304 nm, and emission

detected at 620 nm, and 665 nm.

TR-FRET ParkinW403A Autoubiquitination: Serial

dilutions of ParkinW403A from 50nM to 6.25nM mixed

with UBA1, UBE2D3, trf-Ub mix. Reaction was

initiated with addition of Mg-ATP, excited at 304 nm,

and emission detected at 620 nm, and 665 nm.

300nM wt-Parkin shown for comparison.

In this report, we show the capabilities of a novel

TR-FRET based platform for probing the activities

of E3 ubiquitin ligases and deubiquitinating

enzymes in real-time and endpoint systems. This

technology fills a longstanding gap in the ability of

researchers to conduct drug discovery with

simple assays tailored for the ubiquitin

proteasome system. Unlike existing technologies,

our platform does not rely on secondary

detection methods such as antibodies. We

de o st ate this platfo ’s o ust ess ith assa s o o l e hi iti g Z’ ≥0.8, a d sho its adaptability to miniaturized formats, making it

ideal for high-throughput screening experiments.

We further provide a proof-of-concept for the

s ste ’s utilit a d si pli it i aki g Ki

determinations, and demonstrate that the

platform holds its weight against a common DUB

substrate, Ubiquitin Rhodamine 110.

0 2 0 4 0 6 0 8 0 1 0 0

-1 0 0

-5 0

0

U b iq u it in a t e d M D M 2 v s . U S P 2 C D T it r a t io n

U S P 2 C D (n M )

Ra

te (

de

lta

)(s

ec

-1)

References

0 1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0

1 0 0 0

2 0 0 0

3 0 0 0

M D M 2 A u t o u b iq u it in a t io n

t im e (m in )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

2 0 0 n M

1 0 0 n M

5 0 n M

2 5 n M

1 2 .5 n M

1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0 .0

0 .5

1 .0

M D M 2 A u t o u b iq u it in a t io n Z '

t im e (m in )

Es

tim

ate

d Z

-fa

cto

r

2 0 0 n M

1 0 0 n M

5 0 n M

2 5 n M

1 2 .5 n M

0 1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0

2 0 0

4 0 0

6 0 0

8 0 0

1 0 0 0

X IA P A u t o u b iq u it in a t io n

t im e (m in )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

3 5 0 n M

1 7 5 n M

8 7 .5 n M

4 3 .7 5 n M

1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0 .0

0 .5

1 .0

X IA P A u t o u b iq u it in a t io n Z '

t im e (m in )

Es

tim

ate

d Z

-fa

cto

r

3 5 0 n M

1 7 5 n M

8 7 .5 n M

4 3 .7 5 n M

0 1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0

5 0 0

1 0 0 0

IT C H A u t o u b iq u it in a t io n

t im e (m in )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

1 3 0 n M

6 5 n M

3 2 .5 n M

1 6 .2 5 n M

8 .1 2 5 n M

1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0 .0

0 .5

1 .0

IT C H A u t o u b iq u it in a t io n Z '

t im e (m in )

Es

tim

ate

d Z

-fa

cto

r

1 3 0 n M

6 5 n M

3 2 .5 n M

1 6 .2 5 n M

8 .1 2 5 n M

0 1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0

5 0 0

1 0 0 0

1 5 0 0

P a r k inW 4 0 3 A

A u t o u b iq u it in a t io n

t im e (m in )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

5 0 n M

2 5 n M

1 2 .5 n M

6 .2 5 n M

3 0 0 n M w t P a rk in

1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0 .0

0 .5

1 .0

P a r k inW 4 0 3 A

A u t o u b iq u it in a t io n Z '

t im e (m in )

Es

tim

ate

d Z

-fa

cto

r

5 0 n M

2 5 n M

1 2 .5 n M

6 .2 5 n M

3 0 0 n M w t P a rk in

0 1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0 1 6 5 1 8 0

0

1 0 0 0

2 0 0 0

3 0 0 0

N E D D 4 A u to u b iq u it in a t io n

t im e (m in )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

5 0 n M

2 5 n M

1 2 .5 n M

6 .2 5 n M

3 .1 2 5 n M

1 5 3 0 4 5 6 0 7 5 9 0 1 0 5 1 2 0 1 3 5 1 5 0

0 .0

0 .5

1 .0

N E D D 4 A u t o u b iq u it in a t io n Z '

t im e (m in )

Es

tim

ate

d Z

-fa

cto

r

5 0 n M

2 5 n M

1 2 .5 n M

6 .2 5 n M

3 .1 2 5 n M

2 0 0 1 0 0 5 0 2 5 1 2 .5

0

5 0 0

1 0 0 0

1 5 0 0

2 0 0 0

2 5 0 0

N e d d 4 A u t o -U b 3 0 m in

N E D D 4 (n M )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

2 0 0 1 0 0 5 0 2 5 1 2 .5

0

5 0 0

1 0 0 0

1 5 0 0

2 0 0 0

M D M 2 A u t o -U b 3 0 m in

M D M 2 (n M )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

wt

Pa

rkin

1 3 0 6 5 3 2 .5 1 6 .2 5 8 .1 2 5

0

2 0 0

4 0 0

6 0 0

8 0 0

1 0 0 0

IT C H A u t o -U b 3 0 m in

IT C H (n M )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

3 5 0 1 7 5 8 7 .5 4 3 .7 5

0

2 0 0

4 0 0

6 0 0

8 0 0

X IA P A u t o -U b 1 5 m in

X IA P (n M )

% S

ign

al:

Ba

ck

gr

ou

nd

(r

S-r

B/

rB

)x1

00

%

0 .0 0 1 0 .0 1 0 .1 1 1 0

0 .0

0 .5

1 .0

U b -A ld e h y d e T it r a t io n & 2 5 0 n M U S P 2 C D

U b iq u it in -A ld e h y d e (M )

No

rm

ali

ze

d U

SP

2c

d A

cti

vit

y

IC 5 0 = 0 .1 5 2M

M D M 2 -U B n

U B - R H 1 1 0

IC 5 0 = 0 .1 7 5M

Amerik, A. Y., & Hochstrasser, M. (2004). Mechanism and

function of deubiquitinating enzymes. Biochimica et

Biophysica Acta (BBA)-Molecular Cell Research, 1695(1),

189-207.

Koyano, F., Okatsu, K., Kosako, H., Tamura, Y., Go, E.,

Kimura, M., ... & Endo, T. (2014). Ubiquitin is

phosphorylated by PINK1 to activate parkin. Nature,

510(7503), 162.

Magennis, S. W., Parsons, S., Pikramenou, Z., Corval, A.,

& Woollins, J. D. (1999). Imidodiphosphinate ligands as

antenna units in luminescent lanthanide

complexes. Chemical communications, (1), 61-62.

Zheng, N., & Shabek, N. (2017). Ubiquitin Ligases:

Structure, Function, and Regulation. Annual Review of

Biochemistry, (0).

Powered by TCPDF (www.tcpdf.org)Powered by TCPDF (www.tcpdf.org)Powered by TCPDF (www.tcpdf.org)Powered by TCPDF (www.tcpdf.org)