ArticlePDF Available

Gelatin degradation assay reveals MMP-9 inhibitors and function of O-glycosylated domain

Authors:

Abstract and Figures

To establish a novel, sensitive and high-throughput gelatinolytic assay to define new inhibitors and compare domain deletion mutants of gelatinase B/matrix metalloproteinase (MMP)-9. Fluorogenic Dye-quenched (DQ)™-gelatin was used as a substrate and biochemical parameters (substrate and enzyme concentrations, DMSO solvent concentrations) were optimized to establish a high-throughput assay system. Various small-sized libraries (ChemDiv, InterBioScreen and ChemBridge) of heterocyclic, drug-like substances were tested and compared with prototypic inhibitors. First, we designed a test system with gelatin as a natural substrate. Second, the assay was validated by selecting a novel pyrimidine-2,4,6-trione (barbiturate) inhibitor. Third, and in line with present structural data on collagenolysis, it was found that deletion of the O-glycosylated region significantly decreased gelatinolytic activity (k(cat)/k(M) ± 40% less than full-length MMP-9). The DQ™-gelatin assay is useful in high-throughput drug screening and exosite targeting. We demonstrate that flexibility between the catalytic and hemopexin domain is functionally critical for gelatinolysis.
Content may be subject to copyright.
Gelatin degradation assay reveals MMP-9 inhibitors and
function of O-glycosylated domain
Jennifer Vandooren, Nathalie Geurts, Erik Martens, Philippe E Van den Steen, Steven De Jonghe, Piet Herdewijn,
Ghislain Opdenakker
Jennifer Vandooren, Nathalie Geurts, Erik Martens, Philippe
E Van den Steen, Ghislain Opdenakker, Laboratory of Im-
munobiology, Rega Institute for Medical Research, University
of Leuven, Minderbroederstraat 10, Leuven B-3000, Belgium
Steven De Jonghe, Piet Herdewijn, Medicinal Chemistry,
Rega Institute for Medical Research, University of Leuven,
Minderbroederstraat 10, Leuven B-3000, Belgium
Author contributions: Vandooren J performed the majority
of the experiments and wrote the majority of the manuscript;
Geurts N, Martens E and Van den Steen PE performed the ex-
periments and assisted in writing the manuscript; De Jonghe
S and Herdewijn P provided the ChemBridge, ChemDiv and
InterBioScreen compound library and were involved in study
design; Opdenakker G co-ordinated the study and was involved
in study design and writing and editing the manuscript.
Supported by A postdoctoral fellow of the Belgian Fund for
Scientic Research (F.W.O. Vlaanderen) (Van den Steen PE); A
research assistant of the F.W.O. Vlaanderen (Geurts N)
Correspondence to: Ghislain Opdenakker, MD, PhD, Profes-
sor, Laboratory of Immunobiology, Rega Institute for Medical
Research, University of Leuven, Minderbroederstraat 10, Leuven
B-3000, Belgium. ghislain.opdenakker@rega.kuleuven.be
Telephone: +32-16-337341 Fax: +32-16-337340
Received: September 17, 2010 Revised: November 18, 2010
Accepted: November 25, 2010
Published online: January 26, 2011
Abstract
AIM: To establish a novel, sensitive and high-through-
put gelatinolytic assay to dene new inhibitors and com-
pare domain deletion mutants of gelatinase B/matrix
metalloproteinase (MMP)-9.
METHODS: Quenched Dye-quenched (DQ)™-gelatin
was used as a substrate and biochemical parameters
(substrate and enzyme concentrations, DMSO solvent
concentrations) were optimized to establish a high-
throughput assay system. Various small-sized libraries
(ChemDiv, InterBioScreen and ChemBridge) of hetero-
cyclic, drug-like substances were tested and compared
with prototypic inhibitors.
RESULTS: First, we designed a test system with gelatin
as a natural substrate. Second, the assay was validated
by selecting a novel pyrimidine-2,4,6-trione (barbitu-
rate) inhibitor. Third, and in line with present structural
data on collagenolysis, it was found that deletion of the
O-glycosylated region significantly decreased gelatino-
lytic activity (kcat/kM ± 40% less than full-length MMP-9).
CONCLUSION: The DQ™-gelatin assay is useful in
high-throughput drug screening and exosite targeting.
We demonstrate that exibility between the catalytic and
hemopexin domain is functionally critical for gelatinolysis.
© 2011 Baishideng. All rights reserved.
Key words: Exosite inhibitors; Fluorogenic substrate;
Gelatin; High-throughput screening assays; Matrix me-
talloproteinase-9; Substrate specicity
Peer reviewers: Caroline A Owen, MD, PhD, FRCP Edin, As-
sistant Professor of Medicine, Division of Pulmonary and Criti-
cal Care Medicine, Brigham and Women’s Hospital, 75 Francis
Street, 905 Thorn Building, Boston, MA 02115, United States;
Yan Huang, MD, PhD, Associate Professor, Department of
Medicine, Medical University of South Carolina, 114 Doughty
Street, Room 531, Charleston, SC 29403, United States
Vandooren J, Geurts N, Martens E, Van den Steen PE, De
Jonghe S, Herdewijn P, Opdenakker G. Gelatin degradation as-
say reveals MMP-9 inhibitors and function of O-glycosylated
domain. World J Biol Chem 2011; 2(1): 14-24 Available from:
URL: http://www.wjgnet.com/1949-8454/full/v2/i1/14.htm
DOI: http://dx.doi.org/10.4331/wjbc.v2.i1.14
INTRODUCTION
Matrix metalloproteinases (MMPs) constitute a family of
ORIGINAL ARTICLE
World J Biol Chem 2011 January 26; 2(1): 14-24
ISSN 1949-8454 (online)
© 2011 Baishideng. All rights reserved.
Online Submissions: http://www.wjgnet.com/1949-8454ofce
wjbc@wjgnet.com
doi:10.4331/wjbc.v2.i1.14
World Journal of
Biological Chemistry
W J B C
14 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
Vandooren J
et al
. Gelatinase test for structure-function analysis
more than 25 soluble or membrane bound Zn2+-depen-
dent proteases involved in remodeling of the extracellular
matrix, and in regulation of the function of bioactive
molecules. MMPs are secreted as latent pro-enzymes and
become activated after disruption of the coordination
between the cysteine of the propeptide and the catalytic
zinc (Zn2+) in the active site, for example by proteolysis[1].
This process is described as the cysteine switch model[2].
During normal physiological processes, such as embryo-
genesis, vasculogenesis, wound healing and stem cell mo-
bilization, MMP activities are regulated by transcriptional
regulation, activation and by endogenous inhibitors, such
as the tissue inhibitors of metalloproteinases. Disturbance
of this essential balance between proteinases and natural
inhibitors leads to uncontrolled MMP activities which re-
sults in pathological conditions such as tumor progression
and metastasis, inammation, neurodegenerative, cardio-
vascular and autoimmune diseases[3-6].
MMP inhibitors (MMPIs) have been considered as po-
tential therapeutics for diseases in which excess MMP ac-
tivity is detrimental. The MMPIs, all sharing a zinc binding
group, are categorized into various classes[6], such as the
hydroxamate based MMPIs[7] (e.g. batimastat), the non-hy-
droxamate based MMPIs[8] (e.g. SB-3CT), novel MMPIs[6]
(barbiturates), synthetic peptides and pseudopeptides[9]
(e.g. Regasepin 1) and biotechnological and macromo-
lecular inhibitors of MMPs[10] (e.g. REGA-3G12). Bio-
availability and MMP-specicity are major bottlenecks in
designing MMPIs. The limited success of broad spectrum
inhibitors in clinical trials stimulated research towards the
development of highly sensitive assay methods to screen
for specic MMP activities and to search for selective in-
hibitors[6,11,12].
One of the most studied and structurally most com-
plex members of the MMP family is MMP-9 or gelatinase
B. In contrast to the constitutively expressed MMP-2 or
gelatinase A, MMP-9 expression is induced by various
agonists. After neutrophil activation, MMP-9 is released
from preformed granules[13]. Since many disease states, e.g.
acute inammation, autoimmunity and invasive cancer, are
associated with excess gelatinase B activation, this enzyme
is an interesting and important target for inhibition[6,13,14].
Here we describe a novel, fast and highly sensitive
method for the screening of MMP-9 inhibitors. Dye-
quenched (DQ)™-gelatin consists of quenched FITC-
labeled gelatin which, upon gelatinolytic activity, is con-
verted into bright fluorescent peptides. This reaction is
conveniently used for in situ zymography techniques[15] and
the substrate conversion was parametrically studied in this
work. In contrast to all other MMPs, only gelatinases have
a gelatin-binding fibronectin domain[16]. Hence, compared
to the small uorogenic peptide (FP) (7-methoxycoumarin-
4yl)Acetyl-Pro-Leu-Gly-Leu-[3-(2,4-dinitrophenyl)-L-2,3
diaminopropionyl]-Ala-Arg-NH2 described by Knight
et al[17], DQ™-gelatin mimics the natural substrate to mea-
sure (MMP-9/MMP-2) gelatinolytic activity with high
sensitivity. We studied the catalytic parameters of DQ™-
gelatin conversion by human MMP-9, on the basis of which
a high-throughput assay for rapid screening of MMP-9
inhibitors was established. With this assay we screened li-
braries (ChemDiv, InterBioScreen, ChemBridge) of small
molecules for MMP-9 inhibition. Out of 1612 compounds,
5 inhibited MMP-9 by more than 50% at concentrations be-
low 40 μmol/L. The best selected novel MMP-9 inhibitor
was structurally analogous to an already described MMPI,
RO-28-2653, which belongs to the class of pyrimidine-
2,4,6-triones (barbiturates)[18]. Finally, it was demonstrated
that this assay is useful for MMP exosite studies, because de-
letion of the O-glycosylated domain resulted in signicantly
reduced catalysis of DQ™-gelatin, in comparison with the
activities of the intact MMP-9/gelatinase B.
MATERIALS AND METHODS
Proteins and reagents
Recombinant human full-length proMMP-9 (MMP-9 FL,
92 kDa) as well as mutants lacking the O-glycosylated do-
main (MMP-9∆OG), or the hemopexin domain (MMP-9∆
Hem), or both the O-glycosylated and hemopexin domain
(MMP-9∆OGHem) and a mutant with a point mutation
in the active site (the catalytic Glu402 is mutated into Ala,
rendering the enzyme inactive) and a point mutation in the
OG domain (Cys468 is mutated into Ala) (MMP-9 MutEC)
were expressed in Sf9 insect cells and puried by gelatin-
Sepharose chromatography. Subsequently, the enzymes
were activated by incubation with the catalytic domain of
stromelysin-1/MMP-3. These techniques were performed
as described previously[13,19,20]. The enzymes were always
used in the assays at a concentration of 0.1 nmol/L unless
mentioned otherwise.
For the fluorogenic gelatin assay, DQ™-gelatin was
purchased from Invitrogen (Carlsbad, CA, USA) and dis-
solved in water at 1 mg/mL. For this assay, all solutions
and dilutions were prepared in assay-buffer (50 mmol/L
Tris-HCl pH 7.6, 150 mmol/L NaCl, 5 mmol/L CaCl2
and 0.01% Tween 20). In all experiments, DQ™-gelatin
was used at a concentration of 2.5 μg/mL, unless men-
tioned otherwise.
The uorogenic DQ
-gelatin assay
The following general protocol was used for the setup
of a uorogenic DQ™- gelatin assay. To a 96-well plate
(Macro-assay plate (chimney, 96-well, black, clear bottom,
Greiner Bio-one, Frickenhausen, Germany), 0.1 nmol/L
(for a nal volume of 100 μL) of the enzyme was added.
For inhibitor tests, the required amount of inhibitor was
added and the plate was incubated for 30 min at 37
(note that in this case the actual concentrations of enzyme
and inhibitor were 1.7 times higher during this incubation
period than in the interval used for substrate conversion).
Subsequently, DQ™-gelatin at a final concentration of
2.5 μg/mL was added. Immediately thereafter, the plate
was placed in the uorescence reader (FL600 Microplate
uorescence reader, Biotek, Highland Park, IL, USA) and
uorescence was measured every 10 min for 2 h at 37
(ex. 485 nm/em. 530 nm). In each experiment, both posi-
tive (no inhibitor) and negative (no enzyme) controls were
included. All data were corrected by subtraction of their
15 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
respective negative controls. Graphs and calculations were
obtained with Prism 5 (GraphPad Software, Inc.). For the
calculation of substrate molarities we used an approxi-
mate molecular weight of 100 000 g/mol.
Optimization of the uorogenic gelatin assay
Concentration ranges of both the full length enzyme (MMP-9
FL) and substrate (DQ™-gelatin) were tested. MMP-9 FL
was serially diluted 1/3 starting with a concentration of
4 nmol/L. The substrate was diluted by 1/2, starting with
40 μg/mL (0.4 μmol/L) DQ™-gelatin. As a negative con-
trol, each substrate dilution, without enzyme and in assay
buffer, was always included as a control for spontaneous
substrate conversion.
Analysis of enzyme kinetics of MMP-9 FL and MMP-9
mutants using DQ
-gelatin
MMP-9 FL, MMP-9 ∆Hem, MMP-9 ∆OG, MMP-9
OGHem and MMP-9 mutEC were used at a concentra-
tion of 1 nmol/L. Each enzyme form was tested at a range
of substrate concentrations (40 μg/mL to 0.075 μg/mL
in a 1/2 dilution series). For each enzyme variant, the cor-
responding kinetic parameters and kinetic graphs were
calculated.
Assay validation with a range of known protease
inhibitors
A random set of available protease inhibitors was tested for
their potential MMP-9 FL inhibition in our uorescent gela-
tin assay. Details of the used inhibitors are summarized in
Table 1. A rst screening was carried out with all compounds
at a concentration of 20 μmol/L. After the initial screening,
the active compounds were tested in a 1/2 dilution series
starting at the highest concentration of 20 μmol/L.
The inuence of DMSO on the uorogenic gelatin assay
In view of the fact that hydrophobic compounds are often
dissolved in DMSO, and 10% DMSO disrupts the interac-
tion between gelatin and MMP-9[13], we evaluated the high-
est concentration of DMSO that may be used without in-
terfering with the test system. Prior to the enzymatic tests,
a series of DMSO dilutions were added to the 96-well
plate containing MMP-9 FL and DQ™-gelatin. Negative
controls were included, containing the used DMSO con-
centration and 2.5 μg/mL DQ™-gelatin.
High-throughput screening for MMP-9 inhibition with the
use of the uorogenic gelatin assay
The compound library: The compound library con-
16 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
Table 1 Set of used protease inhibitors in control experiments
Inhibitor (%
inhibition)
Alternative name MW
(g/mol)
Target Mechanism Ref.
Aprotinin
(5%)
Bovine pancreatic trypsin inhibitor Approxi-
mately
6500
Serine proteases including plasmin,
tissue plasminogen activator,
kallikrein and thrombin
Nonspecic protease inhibitor [33,34]
Batimastat
(94%)
BB-94; [4-(N-hydroxyamino)-2R-isobutyl-
3S-(thiopen-2-ylthiomethyl)succinyl]-L-
phenylalanine-N-methylamide
478 MMP-1, -2, -3, -7 and -9 Peptide backbone similar to
the cleavage site in collagen (=
peptidomimetic inhibitor)
[6,9,35-37]
TACE (MMP IC50 = 10-30 nmol/L)
Benzamidine
(0%)
- 120 Trypsin, plasmin and thrombin Competitive inhibitor [36]
Bestatin (0%) [(2S,3R)-3-amino-2-hydroxy-4-
phenylbutanoyl]-L-leucine, Ubenimex
308 Aminopeptidase N Slow-binding competitive inhibitor [38-40]
Chymostatin
(0%)
- 608 Proteinases including Serine, thiol,
and carboxyl endopeptidases serine
proteinases, chymotrypsin and
Streptomyces griseus proteinase A,
and several cysteine proteinases
Tetrapeptide analogue, formation of a
hemiacetal or hemithioacetal adduct
with the nucleophilic hydroxy or
thiol group of the serine and cysteine
proteinases
[41,42]
E-64d (0%) Aloxistatin, EST, [2S,3S-trans-
(Ethoxycarbonyloxirane-2-carbonyl)-L
leucine-(3-methylbutyl) amide]
342 Specic thiol protease inhibitor such
as papain and cathepsin B
Interaction with active thiol group [43]
EGCG (33%) Epigallocatechin-3-gallate 458 Multiple targets including MMP-2 and
MMP-9 (MMP IC50 = 8-50 μmol/L)
Blocking the activation mechanism of
MMP-2 induced by concanavalin A
[44-47]
Other exact molecular targets remain
unknown
Pefabloc (0%) AEBSF; 4-(2-Aminoethyl)
benzenesulfonyl uoride hydrochloride
239 Serine protease inhibitor Irreversible inhibition by covalent
interaction with the active-site serine
[48,49]
Pepstatin
(0%)
Isovaleryl-L-valyl-L-valyl-4-amino-3-
hydroxy-6-methylheptanoyl-L- alanyl-4-
amino-3-hydroxy-6-methylheptanoic acid
686 Pepsin and gastricsin (acid proteinase
activity)
-[50]
PMSF (0%) Phenylmethylsulfonyl uoride 174 Serine protease/carboxylesterase
inhibitor
Covalent binding to the serine
residue of the catalytic Ser-His-Asp
triad
[51]
SB-3CT (91%) - 306 MMP-2 and MMP-9 (MMP IC50 =
185-290 nmol/L)
Competitive, mechanism-based,
thiirane-opening mechanism
[8,52]
The chemical compounds were drawn with ACD/ChemSketch Freeware Software and the structure of Aprotinin was obtained from the Protein Data Bank
(PDB ID: 3LDI). The inhibition percentages are shown below the compound names, as obtained in the initial screen with the compounds at 20 μmol/L.
MMP: Matrix metalloproteinase.
Vandooren J
et al
. Gelatinase test for structure-function analysis
tained in total 1612 small-molecule compounds (MW ap-
proximately 300 g/mol). 555 were purchased from Chem-
Div (San Diego, CA, USA), 360 from InterBioScreen Ltd.
(Moscow, RUS) and 697 from ChemBridge Corporation
(San Diego, CA, USA). All compounds were first dis-
solved in DMSO (concentration of 10 mmol/L). The
compounds were prediluted in assay buffer.
Initial screening: All compounds were tested at a nal
concentration of 20 μmol/L. For each compound a nega-
tive control was included (the enzyme was replaced by
assay buffer). For each plate a positive enzyme control
was included (no inhibitor but an equivalent amount of
DMSO; 0.2%). The data for each compound were cor-
rected with its negative control and compared with the
positive control, giving a percentage decrease in uores-
cence. The compounds which showed more than 20%
inhibitory activity were tested twice more for corrobora-
tions. Inhibition percentages were calculated based on the
uorescence measurement after 2 h.
Dose response: All active compounds were tested again
but at multiple concentrations (1/2 dilution starting at a
concentration of 40 μmol/L and ending at a concentra-
tion of 0.312 μmol/L). For each compound the IC50 was
calculated and a dose response plot was drawn.
FP assay
If necessary, extra information on catalysis by MMP-9
was obtained by using a second FP substrate; {DNP-
Pro-Cha-Gly-Cys(Me)-His-Ala-Lys(N-Me-Abz)-NH2 (ex.
365 nm/em. 450 nm, MM: 1077.2/EMD/Calbiochem®,
Darmstadt, Germany). This substrate is cleaved to a single
cleavage product, Dnm-Pro-Cha-Gly. It can be used for
the evaluation of MMP-9 inhibitors in a uorescent plate
reader[21 ]. MMP-9 FL was used at a concentration of
1 nmol/L (vs 0.1 nmol/L in the uorogenic gelatin assay)
and the FP was used at a concentration of 10 μg/mL (vs
2.5 μg/mL in the uorogenic gelatin assay). Fluorescence
was measured every 10 min for 2 h with the uorescence
reader (FL600 Microplate fluorescence reader, Biotek,
Highland Park, IL, USA).
RESULTS
Assay optimization and validation
Development of the uorescent gelatin assay: An en-
zyme assay was developed with DQ™-gelatin as substrate.
By using different substrate/enzyme concentrations, we
determined the sensitivity of the assay and the optimal
substrate concentration. Figure 1 shows a 3D surface
representation of the signal (measured uorescence at the
respective enzyme and substrate concentration) to noise
(fluorescence measured in wells only containing DQ™-
gelatin = spontaneous degradation) ratio at variable sub-
strate and enzyme concentrations. At lower enzyme con-
centrations the signal-to-noise ratio dropped signicantly.
Based on a compromise between a good detection signal
and minimal enzyme use, we selected the concentration
of 0.1 nmol/L as the enzyme concentration for further
testing in high-throughput drug screening. The yellow line
(Figure 1A) shows this optimal enzyme concentration and
Figure 1B shows uorescence as a function of gelatinase
B concentration with a fixed substrate concentration of
2.5 μg/mL.
To determine the optimal substrate concentration, we
made similar compromises and dened 2.5 μg/mL DQ™-
gelatin as the optimal substrate concentration. This con-
centration is represented by the red line in Figure 1A and
again in Figure 1C. By using only 2.5 μg/mL substrate,
MMP-9 FL levels below 0.1 nmol/L (corresponding to
920 pg) could still be detected.
Standard: To determine the relationship between uores-
cence and product formation a standard curve was con-
structed. A 1/2 dilution series of the substrate was prepared
and ranged from 10 μg/mL DQ™-gelatin to 0.01 μg/mL
DQ™-gelatin. In one dilution series, 0.2 nmol/L of MMP-9
was added. A negative control for spontaneous degrada-
tion was included. When all substrate was converted to
product, when no more changes in uorescence were ob-
served, the uorescence was measured. By using a linear
regression analysis we determined that the fluorescence
was proportional to the converted substrate concentration
(in μmol/L) (Figure 2). 46 h later, another reading was
done, which showed that fluorescence dropped slightly
with time (less than 6%). With the use of a Wilcoxon
signed rank test we found that the difference between
both graphs was signicant (P = 0.0269).
Enzyme kinetics of MMP-9 FL and MMP-9 mutants
using DQ™-gelatin: MMP-9 FL, MMP-9 ∆Hem, MMP-9
∆OG, MMP-9 ∆OGHem and MutEC activity were tested
using the uorescent gelatin assay. The Michaelis-Men-
ten curves and Vmax and kcat/KM parameters are shown in
Figure 3 and Table 2. Deletion of the Hemopexin or the
Hemopexin and O-glycosylated domain seemed to have
least inuence on the enzyme efciency. kcat/KM was re-
duced by ± 10% (relative to the parameters obtained for
MMP-9 FL). As expected, the inactive MMP-9 MutEC did
not show any signicant activity. Interestingly, the MMP-9
∆OG was less active (kcat/KM ± 40% less efcient) than
the mutant lacking both O-glycosylated and hemopexin
domains, suggesting an important role for the linker (OG)
domain for MMP-9 gelatinolytic activity (vida infra). This
OG-domain is a highly glycosylated and proline-rich
sequence of approximately 64 amino acids. It links the
active site and hemopexin domain, but its exact function
remains elusive[19].
Assay validation with a range of known protease inhib-
itors: Initial screening at 20 μmol/L inhibitor concentration
showed that only SB-3CT, BB-94 and EGCG signicantly
lowered MMP-9 activity (for inhibition percentages, Table 1).
As also shown in Table 1, SB-3CT and BB-94 are two in-
hibitors known for their inhibitory activity against MMPs.
In our assay, BB-94 and SB-3CT impaired MMP-9 FL
gelatinolytic activity in the nmol/L range, with BB-94 be-
17 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
Vandooren J
et al
. Gelatinase test for structure-function analysis
ing the best inhibitor (Figure 4). EGCG impaired MMP-9
gelatinolytic activity in the μmol/L range.
Inuence of DMSO on the uorogenic gelatin assay:
Since most commercially available compound libraries are
dissolved in DMSO, we tested whether DMSO had an
influence on the assay. This was expected, since DMSO
disrupts the binding of MMP-9 to gelatin[13]. Figure 5A
shows the enzyme velocity as a function of DMSO con-
centration. With the used conditions, DMSO signicantly
inhibited the enzyme activity with an IC50 of 56 mmol/L
DMSO. Therefore, we tested different DMSO concen-
trations to define a low concentration at which the net
inhibitory effect could still be measured (Figure 5B). At a
concentration of 44 mmol/L DMSO (0.3% DMSO), the
interference was ± 42% of the signal and at 22 mmol/L
(0.15% DMSO), the DMSO interference was ± 24%.
18 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
2000
1500
1000
500
0
Fluorescence
0.1 nmol/L MMP-9
2.5 μg/mL DQ™-gelatin
0 1 2 4
MMP-9 (nmol/L)
4000
3000
2000
1000
0
Fluorescence
2.5 μg/mL DQ™-gelatin
0.1 nmol/L MMP-9
0 10 20 30 40
DQ™-gelatin (μg/mL)
14
12
10
8
6
4
2
0
Signal/noise
40
20
10
5
2.5
1.25
0.625
0.3125
DQ™-gelatin (μg/mL)
0.000152
0.000457
0.001372
0.004115
0.037037
0.111111
0.333333
0.012346
1.000000
3.000000
MMP-9 (nmol/L)
A
CB
Figure 1 Optimization of enzyme and substrate concentrations. A: 3D surface representation of the signal uorescence divided by the noise uorescence (signal/
noise) as a function of the enzyme [matrix metalloproteinase (MMP)-9 FL] and substrate (DQ™-gelatin) concentration. Data were obtained after an incubation period
of 2 h. The red line represents the signal-to-noise ratio as a function of variable enzyme concentration and at a constant substrate concentration of 2.5 μg/mL. The
yellow line shows the signal-to-noise ratio at variable substrate concentrations and at a constant enzyme concentration of approximately 0.1 nmol/L. These enzyme
and substrate concentrations were chosen for further testing; B: The uorescence signal (light blue surface) and noise uorescence (dark blue surface) under different
enzyme concentrations and at a constant substrate concentration of 2.5 μg/mL is shown; C: The uorescence signal (light blue surface) and noise uorescence (dark
blue surface) under different substrate concentrations and at a constant concentration of 0.1 nmol/L MMP-9 FL is plotted.
6000
4000
2000
0
Fluorescence
Standard (12 h):
y = x48 200 ± 90.25
Standard (46 h):
y = x45 150 ± 108.7
0.00 0.02 0.04 0.06 0.08 0.10
DQ™-gelatin (μmol/L)
Figure 2 Standard curves of the correlations between uorescence and
product (DQ™-gelatin) concentration. The full line represents a linear regres-
sion of uorescence data obtained after 12 h incubation. The dashed line repre-
sents a linear regression of the uorescence data obtained after 46 h. The drop
in uorescence was signicant (P < 0.05). Data represent mean ± SE (n = 32).
Vandooren J
et al
. Gelatinase test for structure-function analysis
Therefore, in subsequent experiments the DMSO concen-
tration was always kept as low as possible. We recommend
keeping the DMSO concentration at 0.2% or lower, if
possible.
High-throughput screening for MMP-9 inhibition with the
use of the uorogenic gelatin assay
Initial screening: The results of the initial screening are
shown in Table 3. Four hundred and fifty seven com-
pounds reduced the uorescence within a range of 1%-10%
compared to the control with an equivalent amount of
DMSO. We assumed that these small percentages were in
the error-range of the assay. One hundred and twenty six
compounds reduced the signal between 11%-20% and 37
compounds inhibited the uorescence signal by more than
20%. The increase in uorescence as a function of incuba-
tion time with and without an active compound is shown
in Figure 6. All assays were replicated three times and in-
hibitory compounds were defined on the basis of thrice
concordant results.
Further testing of active compounds: Out of the 37
MMP-9 inhibitors, 5 showed an IC50 value below 40 μmol/L.
The dose response graphs, IC50s and molecular structures
are shown in Figure 7. The two most active compounds
had an IC50 of 15 μmol/L and 19 μmol/L. One of these
compounds was compound 6994210 (ChemBridge) or
19 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
Table 2 Michaelis-Menten parameters for different enzyme variants
MMP-9 FL MMP-9 ∆Hem MMP-9 ∆OGHem MMP-9 MMP-9 MutEC
Vmax (nmol/L per minute) 3.643 2.686 2.314 1.117 -
kcat/KM (nmol/L per minute) 0.097 0.086 0.088 0.058 -
Goodness of t (R2) 0.9977 0.9900 0.9861 0.9470 0.7096
Difference from MMP-9 FL - P = 0.0207 P = 0.0049 P = 0.0020 P = 0.0010
The corresponding Michaelis-Menten curves are shown in Figure 3. The P-values were calculated with a Wilcoxon signed rank test. The Vmax and KM
values could not be determined for the matrix metalloproteinase (MMP)-9 MutEC.
4
3
2
1
0
Enzyme velocity
(nmol/L product/min)
MMP-9 DOGHemb
MMP-9 DHema
MMP-9 FL
MMP-9 DOGb
MMP-9 MutECb
0 100 200 300 400
DQ™-gelatin (nmol/L)
Figure 3 Enzyme velocity as a function of the amount of substrate
(nmol/L DQ™-gelatin) (at a concentration of 1 nmol/L). Prism 5 (GraphPad
Software, Inc) was used to t the data with the corresponding Michaelis-Menten
curve and to calculate the Vmax and KM values (Table 2). By using a Wilcoxon
signed rank test we determined that all mutants had a significantly different
activity from that of matrix metalloproteinase (MMP)-9 FL (aP < 0.05, bP < 0.01).
The graphs are representative of three independent experiments.
2.0
1.5
1.0
0.5
0.0
Enzyme velocity (nmol/min)
0.0 0.5 1.0 1.5
Log (SB-3CT) (μmol/L)
SB-3CT
2.0
1.5
1.0
0.5
0.0
Enzyme velocity (nmol/min)
0.0 0.5 1.0 1.5
Log (BB-94) (μmol/L)
BB-94
Enzyme velocity (nmol/min)
0.0 0.5 1.0 1.5
Log (EGCG) (μmol/L)
EGCG
2.0
1.8
1.6
1.4
1.2
1.0
Figure 4 Dose-response curves of the inhibitory activities of SB-3CT,
BB-94 and EGCG. With GraphPad prism software, the IC50 of SB-3CT and
BB-94 was predicted to be in the nmol/L range and the IC50 of EGCG in the
μmol/L range. The data points correspond to inhibitor concentrations of: 1.25,
2.5, 5, 10 and 20 μmol/L, respectively. The horizontal line shows the enzyme
velocity in the absence of inhibitor.
Vandooren J
et al
. Gelatinase test for structure-function analysis
20 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
5-[(2-hydroxy-6-methyl-3-quinolinyl)methylene]-2,4,-
(1H,3H,5H)-pyrimidinetrione. Pyrimidine-triones have
already been described as metalloproteinase inhibitors.
They are known for their zinc-chelating activity and
substituents have already been optimized to comparable
inhibitory efciency as batimastat (IC50 = 10 nmol/L for
MMP-2 and IC50 of 12 nmol/L for MMP-9) and specic-
ity for MMP-2 and MMP-9[18]. Therefore, the activity of
compound 6994210 may be caused by its zinc-binding
pyrimidine-trione group. Tochowicz et al[22] described
the interaction of compound RO-206-0222 (a barbitu-
ric acid inhibitor) with the MMP-9 catalytic site (of an
inactive E402Q mutant). This compound is a barbituric
acid derivative with two substituents: a phenoxyphenyl
and a pyrimidine-piperazine and gives a tight binding
in the active site of this MMP-9. The barbiturate ring
chelates the catalytic zinc and orients both substituents
into their respective subsites[22]. Intriguingly, compound
0204-5272 (ChemDiv) or N-[4-(6-methyl-1,3-benzothia-
zol-2-yl)phenyl]tetrahydrothiophene-2-carboxamide did
not show any similarity with existing inhibitors.
Compound 5805026 (ChemBridge) or N-(4-ethoxy-
8-methyl-2-quinazolinyl)guanidine was the third most active
compound (IC50 = 25 μmol/L). Compound STOCK1S-
82005 (InterBioScreen) displayed an IC50 of 27 μmol/L
100
80
60
40
20
0
% Activity
22 mmol/L
44 mmol/L
88 mmol/L
IC50 = 56 mmol/L
-2 -1 0 1
Log nal (DMSO) (mol/L)
Fluorescence
44 mmol/L DMSO
0 100 200 300
t
/min
1200
1000
800
600
400
200
0
Fluorescence
22 mmol/L DMSO
0 100 200 300
t
/min
1200
1000
800
600
400
200
0
Fluorescence
88 mmol/L DMSO
0 100 200 300
t
/min
1200
1000
800
600
400
200
0
DC
BA
Figure 5 The inuence of DMSO on the conversion of DQ™-gelatin into uorogenic gelatin by matrix metalloproteinase-9. A: By using a non-linear t, an
IC50 of 56 mmol/L DMSO (R2 = 0.9867) (horizontal dotted line) was determined. The vertical striped lines represent the concentrations used in panels B, C and D; B:
Inuences of 88 mmol/L (0.6% DMSO), 44 mmol/L (0.3% DMSO) and 22 mmol/L DMSO (0.15% DMSO) on the uorescence changes at different time points. The
solid lines show the uorescence evolutions measured in the absence of DMSO, the striped lines show the uorescence measured in the presence of DMSO at the
indicated concentrations. The vertical dotted lines represent uorescence data measured after 2 h.
Table 3 Results of the initial screening of 1612 compounds
Fluorescence decrease
1%-10% 11%-20% > 20%
ChemDiv (555 compounds) 217 57 4
(Max = 33%)
InterBioScreen (360 compounds) 106 19 18
(Max = 100%)
ChemBridge (697 compounds) 134 50 15
(Max = 100%)
800
600
400
200
0
Fluorescence
20 40 60 80 100 120
t
/min
59% decrease
Figure 6 Typical increase in uorescence (per time unit) between the posi-
tive control catalysis and in the presence of an active compound (Chem-
Bridge, 6994210). The percentage inhibition was measured after 2 h.
Positive control catalysis
Active compound
Vandooren J
et al
. Gelatinase test for structure-function analysis
21 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
Compound 0204-5272 (Chemdiv)
2.0
1.5
1.0
0.5
0.0
Velocity (nmol/min)
Compound 0204-5272 (Chemdiv)
IC50 = 14.73 μmol/L
-1.0 -0.5 0.0 0.5 1.0 1.5 2.0
Log (0204-5272)
Autouorescence
S
N
S
NH
O
H3C
2.0
1.5
1.0
0.5
0.0
8
6
4
2
0
D Fluorescence/min
Compound 6994210 (ChemBridge)
-1.0 -0.5 0.0 0.5 1.0 1.5 2.0
Log (6994210)
Velocity (nmol/min)
Compound 6994210 (ChemBridge)
IC50 = 18.55 μmol/L
-1.0 -0.5 0.0 0.5 1.0 1.5 2.0
Log (6994210)
NHHN
O
O
O
NOH
H3C
2.0
1.5
1.0
0.5
0.0
Velocity (nmol/min)
Compound 5805026 (ChemBridge)
IC50 = 24.83 μmol/L
-1.0 -0.5 0.0 0.5 1.0 1.5 2.0
Log (5805026)
Autouorescence
Compound 5805026 (ChemBridge)
CH3
CH3
O
N
N
NH
NH
NH2
8
6
4
2
0
D Fluorescence/min
Compound STOCK1S-82005 (InterBioScreen)
-1.0 -0.5 0.0 0.5 1.0 1.5 2.0
Log (STOCK1S-82005)
2.0
1.5
1.0
0.5
0.0
Velocity (nmol/min)
Compound STOCK1S-82005 (InterBioScreen)
IC50 = 27.48 μmol/L
-1.0 -0.5 0.0 0.5 1.0 1.5 2.0
Log (STOCK1S-82005)
O
CH3
N
N
N
N
H
F
2.0
1.5
1.0
0.5
0.0
Velocity (nmol/min)
Compound C920-1611 (ChemDiv)
IC50 = 29.99 μmol/L
-1.0 -0.5 0.0 0.5 1.0 1.5 2.0
Log (C920-1611)
Compound C920-1611 (ChemDiv)
Autouorescence
NH
CH3
O
N
N
S
Cl
Figure 7 Dose-response graph, IC50 and molecular structure of the 5 most active compounds (IC50 < 40 μmol/L) on conversion of DQ™-gelatin and a uo-
rescent peptide by matrix metalloproteinase-9. The results obtained with the DQ™-gelatin assay (including the IC50s) are shown in the left column. The chemical
structures are shown in the central column. Data with the uorescent peptide are shown in the right column.
Vandooren J
et al
. Gelatinase test for structure-function analysis
22 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
and com pound C920-1611 (Chem Div) or N -(2, 4 -
dimethylphenyl)-2-[(2-methyl-1,3-benzothiazol-6-yl)sulfo-
nylamino] acetamide had an IC50 of ± 30 μmol/L. For
these compounds, no structural similarity could be found
with existing MMP small-molecule inhibitors.
Inhibitor testing with the use of a small FP substrate:
As a comparison, we used a different assay with a FP
substrate to test the inhibitory potential of our 5 newly
discovered inhibitors. However, 3 of the 5 compounds
(compound 0204-5272, compound 5805026 and com-
pound C920-1611) were autofluorescent at the wave-
lengths required for this substrate (Figure 7 right column).
In addition, no inhibition was detected for the other two
compounds, illustrating the power of our new assay.
DISCUSSION
The DQ™-gelatin substrate was originally introduced for
the fluorometric determination of gelatinolytic activity
of cancer cells in vitro[23] but was, until now, mainly used
for the in situ demonstration of gelatinolytic activity[15,24,25].
Here, we show that the DQ™-gelatin assay is a useful tool
in many ways for the biochemical study of gelatinolysis
of puried proteases e.g. MMP-9. With low amounts of
substrate (2.5 μg/mL) and enzyme (0.1 nmol/L), MMP-9
activity was determined accurately. For comparison, with
the fluorogenic peptide {DNP-Pro-Cha-Gly-Cys(Me)-
His-Ala-Lys(N-Me-Abz)-NH2, the optimal substrate and
enzyme concentrations were 10 μg/mL and 1 nmol/L,
respectively. Although {DNP-Pro-Cha-Gly-Cys(Me)-His-
Ala-Lys(N-Me-Abz)-NH2 was originally described as a
good peptide for high-throughput screening efforts and
has compatible emission and excitation spectra with most
uorescent plate readers[21], the uorescent signal is not as
sensitive and stable as with the DQ™-gelatin substrate.
In addition, the DQ™-gelatin substrate is a ‘natural’
MMP-9 substrate compared to short peptides. MMP-9
cooperatively binds gelatin with its fibronectin domain
and catalytic site, thereby orienting the substrate into the
catalytic site. The fibronectin domain is, therefore, also
essential for the gelatinolytic activity[26]. With the use of
gelatin as a natural substrate, the possibility exists of nd-
ing inhibitors targeting the fibronectin-like domain and
exclusively impairing gelatinolytic activity, without having
major implications on other MMP-9 proteolytic events.
Previous clinical trials with MMPIs have been somewhat
disappointing. One often invoked reason is the lack of
specicity, since most existing MMPIs target the catalytic
site, which is shown to be highly conserved and, there-
fore, similar amongst MMPs. Presently, attention is more
focused on distal surface residues and accessory domains
(called “MMP allosteric sites” or “exosites”) which may
allocate single or sets of MMPs and would, therefore, be
good targets for specic MMP inhibition[16]. Some efforts
in this direction have been made. Inhibitory peptides of
the MMP-2 collagen binding domain (CBD) have been
identified by Xu et al[27]. These peptides were also active
against MMP-9.
The possibility exists that the described inhibitory
effect of DMSO is related to the fibronectin domain
exosite. Indeed, recombinant MMP-2 CBD binds to gela-
tin and this complex dissociates in the presence of 2%
DMSO[28]. Furthermore, 2% DMSO, which corresponds
to 280 mmol/L, signicantly reduced the gelatinolytic ac-
tivity of MMP-2[29]. Our ndings suggest that 2% DMSO
has an even higher inhibitory effect (> 80% decreased
activity) on MMP-9. This difference may be due to the
known fact that MMP-9/gelatin binding (through the -
bronectin domain) is dependent on cooperativity between
the fibronectin type
and type modules, whereas
MMP-2 can bind gelatin without the need of cooperativ-
ity[28]. Also, in accordance with these ndings for MMP-2,
DMSO had no inuence on MMP-9 processing of a small
peptide substrate, suggesting that the CBD is not required
for positioning such short peptide substrates relative to
the active site[29].
A method for high throughput screening of potentially
selective MMP-13 (collagenase) exosite inhibitors was de-
veloped by Lauer-Fields et al[30]. They used a triple-helical
FRET substrate and found 34 active compounds includ-
ing two pyrimidine-trione derivatives and new compounds
which did not target the MMP-13 catalytic site. With the
DQ™-gelatin assay we tested 1612 small-molecule com-
pounds for their potential inhibition of MMP-9 FL gelati-
nolytic activity. We identied ve compounds with an IC50
below 30 μmol/L. One of these compounds (6994210)
was a pyrimidinetrione derivative. Barbiturates have previ-
ously been identied as Zn2+-binders[6]. We did not trace
the other small-molecules in the existing literature, mak-
ing these compounds additional candidates for further
development towards MMPIs. The finding of an exist-
ing MMP-9 zinc binder by using the DQ™-gelatin assay
endorses the suitability of this assay for high-throughput
drug screening. In line with this, we were able to perfectly
distinguish the three known MMP-9 inhibitors (SB-3CT,
BB-94 and EGCG) out of a set of 11 other protease
inhibitors with specificities for various (other) protease
classes.
Besides the above-mentioned application, the DQ™-
gelatin substrate was also useful in fundamental studies
of MMP-9 action. We tested different MMP-9 mutants
(MMP-9 ∆Hem, MMP-9 ∆OG, MMP-9 ∆OGHem and
MMP-9 MutEC) in the DQ™-gelatin assay. The fact that
the MMP-9 ∆OG mutant form was ± 40% less efficient
than the MMP-9 FL or the MMP-9 ∆OGHem form, sug-
gests an important role for the OG-domain in MMP-9 ge-
latinolytic activity. This has been suggested by Rosenblum
et al[31] on the basis of structural data. With the use of
single-molecule imaging statistical analysis and small-
angle X-ray scattering (SAXS), it was shown that MMP-9
FL is much more flexible than MMP-9 ∆OG. The OG
domain thus lends the MMP-9 molecule flexibility, sup-
porting multiple enzyme conformations[31]. With the use
of atomic force microscopy, it was recently shown that
MMP-9 FL can adopt an extended and a contracted con-
formation, addressed by the OG domain. Upon binding
of collagen, MMP-9 changes from the extended into the
Vandooren J
et al
. Gelatinase test for structure-function analysis
23 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
contracted form, thereby using the flexibility of the en-
zyme O-glycosylated domain to nd an appropriate bind-
ing site[32]. Removal of this exible linker may thus result
in a rigid structure which has fewer degrees of freedom
for interaction with the gelatin substrate. Removal of the
OG domain, also results in direct contact between the
catalytic and hemopexin domains. Our data on in vitro
gelatinolysis demonstrate functionally the importance of
the O-glycosylated domain in comparison with the hemo-
pexin domain, and further underline the possibilities of
the development of allosteric inhibitors.
We conclude that the DQ™-gelatin assay is useful in
high-throughput drug screening and exosite studies of
MMPs. The assay is easily applicable in multi-well plates
and the substrate is compatible with emission and excita-
tion spectra on most uorescent plate readers. In addition,
less autofluorescence of the compounds is measured at
these wavelengths. Because of the high resolution of the
assay, only small amounts of enzyme and substrate are
necessary, which implies low costs. Besides the technologi-
cal advancements, this study provides further insights into
the MMP inhibitory role of DMSO mediated through the
fibronectin domain and functionally defines the O-gly-
cosylated domain as a crucial entity for gelatin substrate
catalysis.
COMMENTS
Background
Matrix metalloproteinases (MMPs) are a family of Zn2+-dependent multidomain
enzymes, involved in pathological processes such as acute and chronic inam-
mation (e.g. rheumatoid arthritis and multiple sclerosis), cancer cell invasion
and metastasis, periodontal diseases, liver and lung diseases. Historically, the
MMPs were classified into gelatinases, collagenases, stromelysins, metallo-
elastases, matrilysins and membrane type MMPs (MT-MMPs), partially based
on substrate conversion. Gelatinase A/MMP-2 and Gelatinase B/MMP-9 repre-
sent the gelatinases, having gelatins as natural substrates.
Research frontiers
Several MMP inhibitors (MMPIs) have been developed over the past 20 years.
However, most clinical trials with MMPIs had poor outcomes and severe side-
effects were observed. Many reasons have been postulated for these results,
but one major problem was low selectivity of the used MMPIs. In order to
increase selectivity, inhibitors that target the distal surfaces of MMPs in addi-
tion to the highly conserved catalytic site, may be more promising. New high-
throughput screening assays which enable the identication of exosite inhibitors
are therefore needed. Instead of commonly used small peptide substrates,
we used high molecular weight gelatin in an attempt to mimic macromolecular
interactions in order to probe exosite interactions.
Innovations and breakthroughs
The present study validates Dye-quenched (DQ)™-gelatin, a uorogenic gelatin
substrate, for high-throughput drug screening of MMPIs. The presented assay
is easy, low cost and has a high resolution. In addition, this assay enables the
identication of exosite inhibitors for gelatinases, since DQ™-gelatin mimics the
natural substrate. Our study also stresses the crucial role of the O-glycosylated
domain in gelatin catalysis and provides further insights into how DMSO inhibits
MMP-9 through the bronectin domain.
Applications
The gelatin degradation assay is useful in fundamental studies of gelatinase ac-
tion and is applicable for high-throughput drug screening of MMPIs. It also has
potential for the identication of exosite inhibitors.
Peer review
The experiments have been carefully performed and the manuscript is clearly
written. A few issue need to be addressed before the paper should be pub-
lished.
REFERENCES
1 Ra HJ, Parks WC. Control of matrix metalloproteinase cata-
lytic activity. Matrix Biol 2007; 26: 587-596
2 Van Wart HE, Birkedal-Hansen H. The cysteine switch: a
principle of regulation of metalloproteinase activity with
potential applicability to the entire matrix metalloproteinase
gene family. Proc Natl Acad Sci USA 1990; 87: 5578-5582
3 Nagase H, Visse R, Murphy G. Structure and function of ma-
trix metalloproteinases and TIMPs. Cardiovasc Res 2006; 69:
562-573
4 Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases:
regulators of the tumor microenvironment. Cell 2010; 141:
52-67
5 Sternlicht MD, Werb Z. How matrix metalloproteinases reg-
ulate cell behavior. Annu Rev Cell Dev Biol 2001; 17: 463-516
6 Hu J, Van den Steen PE, Sang QX, Opdenakker G. Matrix
metalloproteinase inhibitors as therapy for inammatory and
vascular diseases. Nat Rev Drug Discov 2007; 6: 480-498
7 Low JA, Johnson MD, Bone EA, Dickson RB. The matrix me-
talloproteinase inhibitor batimastat (BB-94) retards human
breast cancer solid tumor growth but not ascites formation in
nude mice. Clin Cancer Res 1996; 2: 1207-1214
8 Tao P, Fisher JF, Mobashery S, Schlegel HB. DFT studies of
the ring-opening mechanism of SB-3CT, a potent inhibitor of
matrix metalloproteinase 2. Org Lett 2009; 11: 2559-2562
9 Hu J, Fiten P, Van den Steen PE, Chaltin P, Opdenakker
G. Simulation of evolution-selected propeptide by high-
throughput selection of a peptidomimetic inhibitor on a capil-
lary DNA sequencer platform. Anal Chem 2005; 77: 2116-2124
10 Paemen L, Martens E, Masure S, Opdenakker G. Monoclonal
antibodies specic for natural human neutrophil gelatinase B
used for afnity purication, quantitation by two-site ELISA
and inhibition of enzymatic activity. Eur J Biochem 1995; 234:
759-765
11 Tu G, Xu W, Huang H, Li S. Progress in the development of
matrix metalloproteinase inhibitors. Curr Med Chem 2008; 15:
1388-1395
12 Cheng G, Wei L, Xiurong W, Xiangzhen L, Shiguang Z,
Songbin F. IL-17 stimulates migration of carotid artery vas-
cular smooth muscle cells in an MMP-9 dependent manner
via p38 MAPK and ERK1/2-dependent NF-kappaB and AP-1
activation. Cell Mol Neurobiol 2009; 29: 1161-1168
13 Masure S, Proost P, Van Damme J, Opdenakker G. Purifi-
cation and identification of 91-kDa neutrophil gelatinase.
Release by the activating peptide interleukin-8. Eur J Biochem
1991; 198: 391-398
14 Van den Steen PE, Dubois B, Nelissen I, Rudd PM, Dwek
RA, Opdenakker G. Biochemistry and molecular biology of
gelatinase B or matrix metalloproteinase-9 (MMP-9). Crit Rev
Biochem Mol Biol 2002; 37: 375-536
15 Oh LY, Larsen PH, Krekoski CA, Edwards DR, Donovan F,
Werb Z, Yong VW. Matrix metalloproteinase-9/gelatinase B
is required for process outgrowth by oligodendrocytes. J Neu-
rosci 1999; 19: 8464-8475
16 Sela-Passwell N, Rosenblum G, Shoham T, Sagi I. Structural
and functional bases for allosteric control of MMP activities:
can it pave the path for selective inhibition? Biochim Biophys
Acta 2010; 1803: 29-38
17 Knight CG, Willenbrock F, Murphy G. A novel coumarin-
labelled peptide for sensitive continuous assays of the matrix
metalloproteinases. FEBS Lett 1992; 296: 263-266
18 Grams F, Brandstetter H, D'Alò S, Geppert D, Krell HW,
Leinert H, Livi V, Menta E, Oliva A, Zimmermann G, Gram
F, Brandstetter H, D'Alò S, Geppert D, Krell HW, Leinert H,
Livi VMenta E, Oliva A, Zimmermann G. Pyrimidine-2,4,6-
Triones: a new effective and selective class of matrix metal-
loproteinase inhibitors. Biol Chem 2001; 382: 1277-1285
19 Van den Steen PE, Van Aelst I, Hvidberg V, Piccard H, Fiten
P, Jacobsen C, Moestrup SK, Fry S, Royle L, Wormald MR,
Wallis R, Rudd PM, Dwek RA, Opdenakker G. The hemo-
COMMENTS
Vandooren J
et al
. Gelatinase test for structure-function analysis
24 January 26, 2011
|
Volume 2
|
Issue 1
|
WJBC
|
www.wjgnet.com
pexin and O-glycosylated domains tune gelatinase B/MMP-9
bioavailability via inhibition and binding to cargo receptors. J
Biol Chem 2006; 281: 18626-18637
20 Geurts N, Martens E, Van Aelst I, Proost P, Opdenakker G,
Van den Steen PE. Beta-hematin interaction with the hemo-
pexin domain of gelatinase B/MMP-9 provokes autocatalytic
processing of the propeptide, thereby priming activation by
MMP-3. Biochemistry 2008; 47: 2689-2699
21 Bickett DM, Green MD, Berman J, Dezube M, Howe AS,
Brown PJ, Roth JT, McGeehan GM. A high throughput uo-
rogenic substrate for interstitial collagenase (MMP-1) and
gelatinase (MMP-9). Anal Biochem 1993; 212: 58-64
22 Tochowicz A, Maskos K, Huber R, Oltenfreiter R, Dive V,
Yiotakis A, Zanda M, Pourmotabbed T, Bode W, Goettig P.
Crystal structures of MMP-9 complexes with ve inhibitors:
contribution of the exible Arg424 side-chain to selectivity. J
Mol Biol 2007; 371: 989-1006
23 Della Porta P, Soeltl R, Krell HW, Collins K, O'Donoghue M,
Schmitt M, Krüger A. Combined treatment with serine prote-
ase inhibitor aprotinin and matrix metalloproteinase inhibi-
tor Batimastat (BB-94) does not prevent invasion of human
esophageal and ovarian carcinoma cells in vivo. Anticancer
Res 1999; 19: 3809-3816
24 Mook OR, Van Overbeek C, Ackema EG, Van Maldegem F,
Frederiks WM. In situ localization of gelatinolytic activity in
the extracellular matrix of metastases of colon cancer in rat
liver using quenched uorogenic DQ-gelatin. J Histochem Cy-
tochem 2003; 51: 821-829
25 Frederiks WM, Mook OR. Metabolic mapping of proteinase
activity with emphasis on in situ zymography of gelatinases:
review and protocols. J Histochem Cytochem 2004; 52: 711-722
26 Pourmotabbed T. Relation between substrate specicity and
domain structure of 92-kDa type IV collagenase. Ann N Y
Acad Sci 1994; 732: 372-374
27 Xu X, Chen Z, Wang Y, Bonewald L, Steffensen B. Inhibition
of MMP-2 gelatinolysis by targeting exodomain-substrate
interactions. Biochem J 2007; 406: 147-155
28 Steffensen B, Wallon UM, Overall CM. Extracellular matrix
binding properties of recombinant fibronectin type II-like
modules of human 72-kDa gelatinase/type IV collagenase.
High afnity binding to native type I collagen but not native
type IV collagen. J Biol Chem 1995; 270: 11555-11566
29 Xu X, Wang Y, Lauer-Fields JL, Fields GB, Steffensen B. Con-
tributions of the MMP-2 collagen binding domain to gelatin
cleavage. Substrate binding via the collagen binding domain
is required for hydrolysis of gelatin but not short peptides.
Matrix Biol 2004; 23: 171-181
30 Lauer-Fields JL, Minond D, Chase PS, Baillargeon PE,
Saldanha SA, Stawikowska R, Hodder P, Fields GB. High
throughput screening of potentially selective MMP-13 exosite
inhibitors utilizing a triple-helical FRET substrate. Bioorg Med
Chem 2009; 17: 990-1005
31 Rosenblum G, Van den Steen PE, Cohen SR, Grossmann JG,
Frenkel J, Sertchook R, Slack N, Strange RW, Opdenakker
G, Sagi I. Insights into the structure and domain flexibility
of full-length pro-matrix metalloproteinase-9/gelatinase B.
Structure 2007; 15: 1227-1236
32 Rosenblum G, Van den Steen PE, Cohen SR, Bitler A, Brand
DD, Opdenakker G, Sagi I. Direct visualization of protease
action on collagen triple helical structure. PLoS One 2010; 5:
e11043
33 Dietrich W. Aprotinin: 1 year on. Curr Opin Anaesthesiol 2009;
22: 121-127
34 Yang IS, Kim TG, Park BS, Cho KJ, Lee JH, Park Y, Kim KH.
Crystal structures of aprotinin and its complex with sucrose
octasulfate reveal multiple modes of interactions with im-
plications for heparin binding. Biochem Biophys Res Commun
2010; 397: 429-435
35 Davies B, Brown PD, East N, Crimmin MJ, Balkwill FR. A
synthetic matrix metalloproteinase inhibitor decreases tumor
burden and prolongs survival of mice bearing human ovar-
ian carcinoma xenografts. Cancer Res 1993; 53: 2087-2091
36 Markwardt F, Landmann H, Walsmann P. Comparative
studies on the inhibition of trypsin, plasmin, and thrombin by
derivatives of benzylamine and benzamidine. Eur J Biochem
1968; 6: 502-506
37 Macaulay VM, O'Byrne KJ, Saunders MP, Braybrooke JP,
Long L, Gleeson F, Mason CS, Harris AL, Brown P, Talbot
DC. Phase I study of intrapleural batimastat (BB-94), a matrix
metalloproteinase inhibitor, in the treatment of malignant
pleural effusions. Clin Cancer Res 1999; 5: 513-520
38 Scornik OA, Botbol V. Bestatin as an experimental tool in
mammals. Curr Drug Metab 2001; 2: 67-85
39 Suda H, Takita T, Aoyagi T, Umezawa H. The structure of
bestatin. J Antibiot (Tokyo) 1976; 29: 100-101
40 Lkhagvaa B, Tani K, Sato K, Toyoda Y, Suzuka C, Sone S.
Bestatin, an inhibitor for aminopeptidases, modulates the
production of cytokines and chemokines by activated mono-
cytes and macrophages. Cytokine 2008; 44: 386-391
41 Tomkinson NP, Galpin IJ, Beynon RJ. Synthetic analogues of
chymostatin. Inhibition of chymotrypsin and Streptomyces
griseus proteinase A. Biochem J 1992; 286 (Pt 2): 475-480
42 Grinde B, Galpin IJ, Wilby AH, Beynon RJ. Inhibition of he-
patic protein degradation by synthetic analogues of chymo-
statin. J Biol Chem 1983; 258: 10821-10823
43 Hanada K, Tamai M, Yamagishi M, Omura S, Sawada J,
Tanaka I. Isolation and characterization of E-64, a new thiol
protease inhibitor. Agric Biol Chem 1978; 42: 523-528
44 Nagle DG, Ferreira D, Zhou YD. Epigallocatechin-3-gallate
(EGCG): chemical and biomedical perspectives. Phytochemis-
try 2006; 67: 1849-1855
45 Sartor L, Pezzato E, Garbisa S. (-)Epigallocatechin-3-gallate
inhibits leukocyte elastase: potential of the phyto-factor in
hindering inammation, emphysema, and invasion. J Leukoc
Biol 2002; 71: 73-79
46 Garbisa S, Biggin S, Cavallarin N, Sartor L, Benelli R, Albini
A. Tumor invasion: molecular shears blunted by green tea.
Nat Med 1999; 5: 1216
47 Demeule M, Brossard M, Pagé M, Gingras D, Béliveau R.
Matrix metalloproteinase inhibition by green tea catechins.
Biochim Biophys Acta 2000; 1478: 51-60
48 Mintz GR. An irreversible serine protease inhibitor. Biopharm
1993; 6: 34-38
49 Megyeri P, Pabst KM, Pabst MJ. Serine protease inhibitors
block priming of monocytes for enhanced release of superox-
ide. Immunology 1995; 86: 629-635
50 Barrett AJ, Dingle JT. The inhibition of tissue acid proteinases
by pepstatin. Biochem J 1972; 127: 439-441
51 De Vendittis E, Ursby T, Rullo R, Gogliettino MA, Masullo M,
Bocchini V. Phenylmethanesulfonyl fluoride inactivates an
archaeal superoxide dismutase by chemical modication of a
specic tyrosine residue. Cloning, sequencing and expression
of the gene coding for Sulfolobus solfataricus superoxide dis-
mutase. Eur J Biochem 2001; 268: 1794-1801
52 Bannikov GA, Lakritz J, Premanandan C, Mattoon JS, Abra-
hamsen EJ. Kinetics of inhibition of puried bovine neutro-
phil matrix metalloproteinase 9 by low-molecular-weight
inhibitors. Am J Vet Res 2009; 70: 633-639
S- Editor Cheng JX L- Editor Webster JR E- Editor Zheng XM
Vandooren J
et al
. Gelatinase test for structure-function analysis
... To analyze the antiwrinkle or antiaging activity of CALE, we measured its collagenase, elastase, and matrix metalloprotein-1 (MMP-1) activities. To measure collagenase activity, we used the fluorogenic dye-quenched (DQ) gelatin method of Vandooren et al. (2011) [57], with modifications. In brief, CALE at different concentrations was mixed with 1 unit/mL collagenase and 15 µg/mL DQ gelatin and then reacted for 20 min. ...
... To analyze the antiwrinkle or antiaging activity of CALE, we measured its collagenase, elastase, and matrix metalloprotein-1 (MMP-1) activities. To measure collagenase activity, we used the fluorogenic dye-quenched (DQ) gelatin method of Vandooren et al. (2011) [57], with modifications. In brief, CALE at different concentrations was mixed with 1 unit/mL collagenase and 15 µg/mL DQ gelatin and then reacted for 20 min. ...
Article
Full-text available
Several studies have explored the biological activities of Citrus aurantium flowers, fruits, and seeds, but the bioactivity of C. aurantium leaves, which are treated as waste, remains unclear. Thus, this study developed a pilot-scale ultrasonic-assisted extraction process using the Box–Behnken design (BBD) for the optimized extraction of active compounds from C. aurantium leaves, and their antityrosinase, antioxidant, antiaging, and antimicrobial activities were evaluated. Under optimal conditions in a 150× scaleup configuration (a 30 L ultrasonic machine) of a pilot plant, the total phenolic content was 69.09 mg gallic acid equivalent/g dry weight, which was slightly lower (3.17%) than the theoretical value. The half maximal inhibitory concentration of C. aurantium leaf extract (CALE) for 2,2-diphenyl-1-picrylhydrazyl–scavenging, 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)–scavenging, antityrosinase, anticollagenase, antielastase and anti-matrix metalloprotein-1 activities were 123.5, 58.5, 181.3, 196.4, 216.3, and 326.4 mg/L, respectively. Moreover, the minimal inhibitory concentrations for bacteria and fungi were 150–350 and 500 mg/L, respectively. In total, 17 active compounds were detected in CALE—with linalool, linalyl acetate, limonene, and α-terpineol having the highest concentrations. Finally, the overall transdermal absorption and permeation efficiency of CALE was 95.9%. In conclusion, our CALE demonstrated potential whitening, antioxidant, antiaging, and antimicrobial activities; it was also nontoxic and easily absorbed into the skin as well as inexpensive to produce. Therefore, it has potential applications in various industries.
... The collagenase activity was measured using a modified fluorogenic DQ-gelatin assay using the method reported by Vandooren et al. (2011) [24]. The collagenase activity was determined by measuring the absorbance at an excitation wavelength of 485 nm and an emission wavelength of 528 nm by using a Synergy 2 microplate reader (BioTek Instruments, Santa Clara, CA, USA). ...
... The collagenase activity was measured using a modified fluorogenic DQ-gelatin assay using the method reported by Vandooren et al. (2011) [24]. The collagenase activity was determined by measuring the absorbance at an excitation wavelength of 485 nm and an emission wavelength of 528 nm by using a Synergy 2 microplate reader (BioTek Instruments, Santa Clara, CA, USA). ...
Article
Full-text available
Chenopodium formosanum Koidz (CF) is an indigenous cereal plant of Taiwan. Its high content of secondary metabolites and nutrients has attracted attention for its use in skin care products and functional foods. However, most studies have focused on the extract of CF seeds, which are relatively expensive, and none have investigated the effects of combining extraction and fermentation. In this study, we evaluated the utility of using extracts of different parts of CF, i.e., the leaves, stems, and unhulled and hulled seeds. We first made aqueous, ethanolic, methanolic, and ethyl acetate extracts of the four parts. After assessing their biological activities, we selected only unfermented and fermented CF leaf methanolic extracts for subsequent analysis. None of the concentrations of fermented CF leaf extract (≤400 mg/L) were cytotoxic, and all exhibited antioxidative, anti-inflammatory, antimicrobial, skin-whitening, moisturizing, and antiaging activities. The concentrations of protocatechuic acid, epicatechin, gallic acid, and quercetin increased the most after fermentation. Therefore, they were subjected to a molecular docking analysis, which revealed that quercetin and epicatechin may contribute the most to skin-whitening and antiaging properties, respectively. In conclusion, fermented CF leaf methanolic extracts can be useful as a functional ingredient in health foods, botanical drugs, and cosmetic products.
... However, it was not possible to determine the molar concentration of the enzyme they used, because they did not specify the final reaction volumes. As another reference to this finding, in the study published by Vandooren et al. [52], different compounds were evaluated using the DQ-gelatin assay. The authors identified the three most efficient inhibitors of MMP-9: a phenylalanine derivative (batimastat), an aromatic ether compound (SB-3CT), and the polyphenol epigallocatechin-3-gallate (EGCC). ...
Article
Full-text available
Chronic obstructive pulmonary disease (COPD) is comprised of histopathological alterations such as pulmonary emphysema and peribronchial fibrosis. Matrix metalloproteinase 9 (MMP-9) is one of the key enzymes involved in both types of tissue remodeling during the development of lung damage. In recent studies, it was demonstrated that deflamin, a protein component extracted from Lupinus albus, markedly inhibits the catalytic activity of MMP-9 in experimental models of colon adenocarcinoma and ulcerative colitis. Therefore, in the present study, we investigated for the first time the biological effect of deflamin in a murine COPD model induced by chronic exposure to ozone. Ozone exposure was carried out in C57BL/6 mice twice a week for six weeks for 3 h each time, and the treated group was orally administered deflamin (20 mg/kg body weight) after each ozone exposure. The histological results showed that deflamin attenuated pulmonary emphysema and peribronchial fibrosis, as evidenced by H&E and Masson’s trichrome staining. Furthermore, deflamin administration significantly decreased MMP-9 activity, as assessed by fluorogenic substrate assay and gelatin zymography. Interestingly, bioinformatic analysis reveals a plausible interaction between deflamin and MMP-9. Collectively, our findings demonstrate the therapeutic potential of deflamin in a COPD murine model, and suggest that the attenuation of the development of lung tissue damage occurs by deflamin-regulated MMP-9 catalytic activity.
... Two substrates were selected for use, namely fluorescently labelled gelatin and collagen type I, with the latter being the most relevant for studying venom-induced cytotoxicity by representing an ECM component. Both polymers are already being used to study non-venom proteases [37][38][39][40][41][42]. ...
Article
Full-text available
The cytotoxicity caused by snake venoms is a serious medical problem that greatly contributes to the morbidity observed in snakebite patients. The cytotoxic components found in snake venoms belong to a variety of toxin classes and may cause cytotoxic effects by targeting a range of molecular structures, including cellular membranes, the extracellular matrix (ECM) and the cytoskeleton. Here, we present a high-throughput assay (384-well plate) that monitors ECM degradation by snake venom toxins via the application of fluorescent versions of model ECM substrates, specifically gelatin and collagen type I. Both crude venoms and fractionated toxins of a selection of medically relevant viperid and elapid species, separated via size-exclusion chromatography, were studied using the self-quenching, fluorescently labelled ECM–polymer substrates. The viperid venoms showed significantly higher proteolytic degradation when compared to elapid venoms, although the venoms with higher snake venom metalloproteinase content did not necessarily exhibit stronger substrate degradation than those with a lower one. Gelatin was generally more readily cleaved than collagen type I. In the viperid venoms, which were subjected to fractionation by SEC, two (B. jararaca and C. rhodostoma, respectively) or three (E. ocellatus) active proteases were identified. Therefore, the assay allows the study of proteolytic activity towards the ECM in vitro for crude and fractionated venoms.
... The MMP9 protein contained a conserved Pro-Arg-Cys-Gly-Val/ASN-PRO-ASP (PRCGV/NDP) sequence immediately following the signal peptide (aa 1-21), and the conserved cysteine residue in that sequence has been found to play a crucial role in the enzymatic activation of most MMPs [30]. The MMP protein also contained three nearly identical fibronectin repeats (FN2), whose function is to promote the degradation of large or gelatinous substrates, such as elastin and gelatine [31]. In addition, the haemagglutinin domain of the ToMMP9 protein shared only 25-33% sequence similarity with the haemagglutinin domain of other members of the matrix metalloproteinase family [32], illustrating its special position in the family. ...
Article
Full-text available
The MMPs are endogenous proteolytic enzymes that require zinc and calcium as cofactors. MMP9 is one of the most complex matrix metalloproteinases in the gelatinase family and has many biological functions. In mammals, mmp9 is thought to be closely associated with cancer. However, studies in fish have rarely been reported. In this study, to understand the expression pattern of the ToMMP9 gene and its association with the resistance of Trachinotus ovatus to Cryptocaryon irritans, the sequence of the MMP9 gene was obtained from the genome database. The expression profiles were measured by qRT–PCR, the SNPs were screened by direct sequencing, and genotyping was performed. The ToMMP9 gene contained a 2058 bp ORF encoding a putative amino acid sequence of 685 residues. The homology of the ToMMP9 in teleosts was more than 85%, and the genome structure of ToMMP9 was conserved in chordates. The ToMMP9 gene was expressed in different tissues of healthy individuals and was highly expressed in the fin, the gill, the liver and the skin tissues. The ToMMP9 expression in the skin of the infected site and its adjacent sites increased significantly after C. irritans infection. Two SNPs were identified in the ToMMP9 gene, and the SNP (+400A/G) located in the first intron was found to be significantly associated with the susceptibility/resistance to C. irritans. These findings suggest that ToMMP9 may play an important role in the immune response of T. ovatus against C. irritans.
... The gelatinolytic activity of matrix metalloproteases was determined in the conditioned medium from brain endothelial cell cultures from 2-mo-old Lrp1 lox/lox ; Tie2-Cre mice and Lrp1 lox/lox littermate controls using DQ-Gelatin (D-12054; Invitrogen) assay as reported (Vandooren et al., 2011). Four independent primary mouse brain endothelial cell cultures (200,000 cells/well, four wells per culture) were used for all studied conditions. ...
Article
Full-text available
The low-density lipoprotein receptor–related protein 1 (LRP1) is an endocytic and cell signaling transmembrane protein. Endothelial LRP1 clears proteinaceous toxins at the blood–brain barrier (BBB), regulates angiogenesis, and is increasingly reduced in Alzheimer’s disease associated with BBB breakdown and neurodegeneration. Whether loss of endothelial LRP1 plays a direct causative role in BBB breakdown and neurodegenerative changes remains elusive. Here, we show that LRP1 inactivation from the mouse endothelium results in progressive BBB breakdown, followed by neuron loss and cognitive deficits, which is reversible by endothelial-specific LRP1 gene therapy. LRP1 endothelial knockout led to a self-autonomous activation of the cyclophilin A–matrix metalloproteinase-9 pathway in the endothelium, causing loss of tight junctions underlying structural BBB impairment. Cyclophilin A inhibition in mice with endothelial-specific LRP1 knockout restored BBB integrity and reversed and prevented neuronal loss and behavioral deficits. Thus, endothelial LRP1 protects against neurodegeneration by inhibiting cyclophilin A, which has implications for the pathophysiology and treatment of neurodegeneration linked to vascular dysfunction.
Article
Full-text available
A long-standing challenge in skeletal tissue engineering is to reconstruct a three-dimensionally (3D) interconnected bone cell network in vitro that mimics the native bone microarchitecture. While conventional hydrogels are extensively used in studying bone cell behavior in vitro, current techniques lack the precision to manipulate the complex pericellular environment found in bone. The goal of this study is to guide single bone cells to form a 3D network in vitro via photosensitized two-photon ablation of microchannels in gelatin methacryloyl (GelMA) hydrogels. A water-soluble two-photon photosensitizer (P2CK) was added to soft GelMA hydrogels to enhance the ablation efficiency. Remarkably, adding 0.5 mM P2CK reduced the energy dosage threshold five-fold compared to untreated controls, enabling more cell-compatible ablation. By employing low-energy ablation (100 J/cm2) with a grid pattern of 1 µm wide and 30 µm deep microchannels, we induced dendritic outgrowth in human mesenchymal stem cells (hMSC). After 7 days, the cells successfully utilized the microchannels and formed a 3D network. Our findings reveal that cellular viability after low-energy ablation was comparable to unablated controls, whereas high-energy ablation (500 J/cm2) resulted in 42% cell death. Low-energy grid ablation significantly promoted network formation and >40 µm long protrusion outgrowth. While the broad-spectrum matrix metalloproteinase inhibitor (GM6001) reduced cell spreading by inhibiting matrix degradation, cells invaded the microchannel grid with long protrusions. Collectively, these results emphasize the potential of photosensitized two-photon hydrogel ablation as a high-precision tool for laser-guided biofabrication of 3D cellular networks in vitro.
Article
Chlorotoxin, a scorpion venom-derived 36-residue miniprotein, binds to and is taken up selectively by glioblastoma cells. Previous studies provided controversial results concerning target protein(s) of CTX. These included: CLC3 chloride channel, matrix metalloproteinase 2 (MMP-2), regulators of MMP-2, annexin A2 (ANX2) and neuropilin 1 (NRP1). The aim of this study was to clarify which of the proposed binding partners can really interact with CTX using biochemical methods and recombinant proteins. For this purpose, we established two new binding assays based on anchoring the tested proteins to microbeads and quantifying the binding of CTX by flow cytometry. Screening of His-tagged proteins anchored to cobalt-coated beads indicated strong interaction of CTX with MMP-2 and NRP1, whereas binding to ANX2 was not confirmed. The same results were obtained with fluorophore-labeled CTX and CTX-displaying phages. Affinity of CTX to MMP-2 and NRP1 was assessed by the 'Ig-coated bead' test, in which the proteins were anchored to beads by specific antibodies. This assay yielded highly reproducible data using both direct titration and displacement approach. The affinities of labeled and unlabeled CTX appeared to be similar for both MMP-2 and NRP1 with estimated KD values of 0.5-0.7 μM. Contrary to previous reports, we found that CTX does not inhibit the activity of MMP-2 and that not only CTX with free carboxyl end but also with carboxamide terminal binds to NRP1. We conclude that the presented robust assays could also be applied for affinity improving studies of CTX to its genuine targets using phage display libraries.
Article
Full-text available
The physiological activity of the 50% ethanolic extract of Citrus aurantium flower before and after fermentation was investigated in this study. C. aurantium flowers grown in Taiwan were extracted using 100% methanol or 50% ethanol and then fermented by one of six microbes: four species of lactic acid bacteria (Bifidobacterium bifidum, Bifidobacterium lactis, Lactobacillus acidophilus, and Lactobacillus brevis) anaerobically cultivated in MRS broth and two species of mold (Aspergillus oryzae and Aspergillus niger) aerobically cultivated in potato dextrose broth. The 50% ethanolic extract of C. aurantium flowers exhibited higher tyrosinase inhibition (IC50: 200.8 ± 11.6 mg/L) and antioxidative activity than did a 100% methanolic extract (IC50: 274.1 ± 15.7 mg/L). The 50% ethanolic extract fermented by L. brevis (L. brevis–fermented extract) exhibited the highest yield (86.2% ± 1.2%) and physiological activity. The L. brevis–fermented extract exhibited over 5.2-, 13.5-, 12.5-, 3.17-, and 4.29-fold higher antityrosinase activity, antioxidative activity, antibacterial activity, total flavonoid content, and antiwrinkle activity than did the unfermented extract. The L. brevis–fermented extract can be considered safe because it exerted no toxic effect on CCD-966SK or HEMn cells at concentrations of 400 and 200 mg/L, respectively. The fermented extract (40 mg/L) inhibited melanin formation, reducing it to 50.8% ± 2.3%. Furthermore, the L. brevis–fermented extract exhibited excellent antiaging and antiwrinkle activity, as determined from MMP-1, MMP-2, elastase, and collagenase activity. The improvement in physiological characteristics, especially the considerable formation of neohesperidin, is mainly attributable to biosynthesis or biotransformation by L. brevis during fermentation. In conclusion, the 50% ethanolic extract of C. aurantium flowers fermented with L. brevis can be used in cosmetics applications aiming for skin-whitening or antiwrinkle properties.
Article
Full-text available
Scars composed of fibrous connective tissues are natural consequences of injury upon incisional wound healing in soft tissues. Hydrogels that feature a sustained presentation of immunomodulatory cytokines are known to modulate wound healing. However, existing immunomodulatory hydrogels lack interconnected micropores to promote cell ingrowth. Other limitations include invasive delivery procedures and harsh synthesis conditions that are incompatible with drug molecules. Here, hybrid nanocomposite microgels containing interleukin‐10 (IL‐10) are reported to modulate tissue macrophage phenotype during wound healing. The intercalation of laponite nanoparticles in the polymer network yields microgels with tissue‐mimetic elasticity (Young's modulus in the range of 2–6 kPa) and allows the sustained release of IL‐10 to promote the differentiation of macrophages toward proregenerative phenotypes. The porous interstitial spaces between microgels promote fibroblast proliferation and fast trafficking (an average speed of ≈14.4 µm h⁻¹). The incorporation of hyaluronic acid further enhances macrophage infiltration. The coculture of macrophages and fibroblasts treated with transforming growth factor‐beta 1 resulted in a twofold reduction in collagen‐I production for microgels releasing IL‐10 compared to the IL‐10 free group. The new microgels show potential toward regenerative healing by harnessing the antifibrotic behavior of host macrophages.
Article
Full-text available
Oligodendrocytes (OLs) extend processes to contact axons as a prerequisite step in myelin formation. As the OL processes migrate toward their axonal targets, they modify adhesion to their substrate, an event that may be regulated by matrix metalloproteinases (MMPs). In the mouse optic nerve, MMP-9/gelatinase B increases during myelin formation. Although tissue inhibitor of metalloproteinase (TIMP)-3 also increases in parallel, the developing optic nerve has focally active MMPs demonstrable by in situ zymography. The distribution of proteolytic activity is similar to that of myelin basic protein, a marker of myelin formation. OLs in culture secrete MMP-9 and express active cell-associated metalloproteinases at the growing tips of their processes. TIMP-1 and a function-perturbing anti-MMP-9 antibody attenuate outgrowth of processes by OLs, indicating a requirement for MMP-9 in process outgrowth. Process reformation is retarded significantly in OLs cultured from MMP-9 null mice, as compared with controls, providing genetic evidence that MMP-9 is necessary for process outgrowth. These data show that MMP-9 facilitates process outgrowth by OLs in vivo and in culture.
Article
Full-text available
Enzymatic processing of extracellular matrix (ECM) macromolecules by matrix metalloproteases (MMPs) is crucial in mediating physiological and pathological cell processes. However, the molecular mechanisms leading to effective physiological enzyme-ECM interactions remain elusive. Only scant information is available on the mode by which matrix proteases degrade ECM substrates. An example is the enzymatic degradation of triple helical collagen II fragments, generated by the collagenase MMP-8 cleavage, during the course of acute inflammatory conditions by gelatinase B/MMP-9. As is the case for many other matrix proteases, it is not clear how MMP-9 recognizes, binds and digests collagen in this important physiological process. We used single molecule imaging to directly visualize this protease during its interaction with collagen fragments. We show that the initial binding is mediated by the diffusion of the protease along the ordered helix on the collagen (3/4) fragment, with preferential binding of the collagen tail. As the reaction progressed and prior to collagen degradation, gelatin-like morphologies resulting from the denaturation of the triple helical collagen were observed. Remarkably, this activity was independent of enzyme proteolysis and was accompanied by significant conformational changes of the working protease. Here we provide the first direct visualization of highly complex mechanisms of macromolecular interactions governing the enzymatic processing of ECM substrates by physiological protease.
Article
A new thiol protease inhibitor, named E–64, was isolated from the extract of a solid culture of Aspergillus japonicus TPR–64 freshly isolated from soil. E–64 was obtained as white needles and the empirical formula was estimated to be C15N5–H27O5 (M. W. 357). This was almost neutral in its electrophoretic behavior and proved to be a specific and strong inhibitor toward thiol protease such as papain and cathepsin B. They combine equimolecularly and irreversibly.
Article
A new thiol protease inhibitor, named E-64, was isolated from the extract of a solid culture of Aspergillus japonicus TPR-64 freshly isolated from soil. E-64 was obtained as white needles and the empirical formula was estimated to be C15N5H27O5 (M. W. 357). This was almost neutral in its electrophoretic behavior and proved to be a specific and strong inhibitor toward thiol protease such as papain and cathepsin B. They combine equimolecularly and irreversibly.
Article
The gene encoding the superoxide dismutase from the hyperthermophilic archaeon Sulfolobus solfataricus (SsSOD) was cloned and sequenced and its expression in Escherichia coli obtained. The chemicophysical properties of the recombinant SsSOD were identical with those of the native enzyme. The recombinant SsSOD possessed a covalent modification of Tyr41, already observed in native SsSOD [Ursby, T., Adinolfi, B.S., Al-Karadaghi, S., De Vendittis, E. & Bocchini, V. (1999) J. Mol. Biol.286, 189–205]. HPLC analysis of SsSOD samples prepared from cells treated or not with phenylmethanesulfonyl fluoride (PhCH2SO2F), a protease inhibitor routinely added during the preparation of cell-free extracts, showed that the modification was caused by PhCH2SO2F. Refinement of the crystal model of SsSOD confirmed that a phenylmethanesulfonyl moiety was attached to the hydroxy group of Tyr41. PhCH2SO2F behaved as an irreversible inactivator of SsSOD; in fact, the specific activity of both native and recombinant enzyme decreased as the percentage of modification increased. The covalent modification caused by PhCH2SO2F reinforced the heat stability of SsSOD. These results show that Tyr41 plays an important role in the enzyme activity and the maintenance of the structural architecture of SsSOD.
Article
The zinc-dependent matrix metalloproteinases (MMPs) belong to a large family of structurally homologous enzymes. These enzymes are involved in a wide variety of biological processes ranging from physiological cell proliferation and differentiation to pathological states associated with tumor metastasis, inflammation, tissue degeneration, and cell death. Controlling the enzymatic activity of specific individual MMPs by antagonist molecules is highly desirable, first, for studying their individual roles, and second as potential therapeutic agents. However, blocking the enzymatic activity with synthetic small inhibitors appears to be an extremely difficult task. Thus, this is an unmet need presumably due to the high structural homology between MMP catalytic domains. Recent reports have recognized a potential role for exosite or allosteric protein regions, distinct from the extended catalytic pocket, in mediating MMP activation and substrate hydrolysis. This raises the possibility that MMP enzymatic and non-enzymatic activities may be modified via antagonist molecules targeted to such allosteric sites or to alternative enzyme domains. In this review, we discuss the structural and functional bases for potential allosteric control of MMPs and highlight potential alternative enzyme domains as targets for designing highly selective MMP inhibitors.
Article
The crystal structures of aprotinin and its complex with sucrose octasulfate (SOS), a polysulfated heparin analog, were determined at 1.7-2.6A resolutions. Aprotinin is monomeric in solution, which associates into a decamer at high salt concentrations. Sulfate ions serve to neutralize the basic amino acid residues of aprotinin to stabilize the decameric aprotinin. Whereas SOS interacts with heparin binding proteins at 1:1 molar ratio, SOS was surprisingly found to induce strong agglutination of aprotinins. Five molecules of aprotinin interact with one molecule of the sulfated sugar, which is stabilized by electrostatic interactions between the positively charged residues of aprotinin and sulfate groups of SOS. The multiple binding modes of SOS with five individual aprotinin molecules may represent the diverse patterns of potential heparin binding to aprotinin, reflecting the interactions of densely packed protein molecules along the heparin polymer.
Article
Extracellular proteolysis mediates tissue homeostasis. In cancer, altered proteolysis leads to unregulated tumor growth, tissue remodeling, inflammation, tissue invasion, and metastasis. The matrix metalloproteinases (MMPs) represent the most prominent family of proteinases associated with tumorigenesis. Recent technological developments have markedly advanced our understanding of MMPs as modulators of the tumor microenvironment. In addition to their role in extracellular matrix turnover and cancer cell migration, MMPs regulate signaling pathways that control cell growth, inflammation, or angiogenesis and may even work in a nonproteolytic manner. These aspects of MMP function are reorienting our approaches to cancer therapy.
Article
SB-3CT is a 2-[(arylsulfonyl)methyl]thiirane that achieves potent inhibition, by a thiirane-opening mechanism, of the MMP2 and MMP9 zinc metalloproteases. The deprotonation mechanism for thiirane opening of SB-3CT and for the opening of its oxirane analogue, both relevant to the inhibition of MMP2, was investigated computationally using the acetate anion as the Brønsted base and in methanol and acetonitrile as solvents. The activation barriers for the reaction show a significant stereoelectronic effect. The lowest energy paths have the breaking C-H bond gauche to both sulfone oxygens and with this C-H bond anti to the breaking C-S bond of the thiirane. The calculated primary isotope effect agrees with experimental data.