Skip to main content
Erschienen in: Molecular Brain 1/2013

Open Access 01.12.2013 | Methodology

Fluorometric immunocapture assay for the specific measurement of matrix metalloproteinase-9 activity in biological samples: application to brain and plasma from rats with ischemic stroke

verfasst von: Kimberly E Hawkins, Kelly M DeMars, Changjun Yang, Gary A Rosenberg, Eduardo Candelario-Jalil

Erschienen in: Molecular Brain | Ausgabe 1/2013

Abstract

Background

Matrix metalloproteinases are important factors in the molecular mechanisms leading to neuronal injury in many neurological disorders. Matrix metalloproteinase (MMP)-9 is up-regulated after cerebral ischemia and neuroinflammation and is actively involved in blood–brain barrier disruption. Current methods of measuring MMP-9 activity, such as gelatin-substrate zymography, are unspecific and arduous. Here we developed an immunocapture assay with high efficiency, specificity, and sensitivity for quantifying endogenously active as well as total MMP-9 activity.

Results

A fluorescence resonance energy transfer (FRET) peptide-based immunocapture assay was developed that enables the accurate assessment of total and active forms of MMP-9 in complex biological samples. The FRET assay demonstrated correct and efficient binding of MMP-9 to a mouse monoclonal MMP-9 antibody and high specificity of the immunocapture antibody for MMP-9. Total and active levels of MMP-9 were measured in rat brain homogenates, plasma, human HT-1080 conditioned media, and RBE4 endothelial cell lysates. The FRET immunocapture assay yielded highly similar results for total MMP-9 activity when compared to gelatin-substrate zymography.

Conclusions

We suggest that the new FRET peptide-based immunocapture assay is a viable replacement of zymography for sensitive and high throughput quantification of MMP-9 activity in biological samples.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1756-6606-6-14) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KEH, GAR, and ECJ conceived and designed research; ECJ led the project; KEH, KMD, CY, and ECJ performed experiments; and KEH and ECJ performed data analysis and wrote the paper. All authors read and approved the final manuscript.
Abkürzungen
MMP
Matrix metalloproteinase
ECM
Extracellular matrix
BBB
Blood–brain barrier
FRET
Fluorescence resonance energy transfer
5-FAM
5-carboxy-fluorescein
APMA
4-aminophenylmercuric acetate
MCAO
Middle cerebral artery occlusion
RFU
Relative fluorescence unit
RBE4
Rat brain endothelial 4
IL-1β
Interleukin-1β
RLU
Relative lysis unit
CCA
Common carotid artery
ECA
External carotid artery
ICA
Internal carotid artery
PVDF
Polyvinylidene fluoride
TBS
Tris-buffered saline.

Background

Matrix metalloproteinases (MMPs) are a family of 23 members of calcium- and zinc-dependent endopeptidases with the ability to degrade extracellular matrix (ECM) proteins, including collagen, fibronectin, laminin, elastin, and proteoglycans [14]. The MMPs are widely studied due to their involvement in several human diseases including cancer, osteoarthritis, inflammation, neurodegeneration, and cerebrovascular diseases.
MMPs are produced as a zymogen (latent pro-MMP devoid of enzymatic activity), but upon activation, they participate in many physiological as well as pathological events. The pro-domain (pro-MMP) contains a cysteine residue that acts as a zinc-coordinating ligand masking the catalytic domain of the protease (‘cysteine switch’) and preventing the zinc atom in the catalytic domain from being available for enzymatic function [5]. Proteolytic cleavage of the pro-domain by proteases (including other MMPs) [6, 7], oxidative modifications of the thiol group on the zinc-binding cysteine (e.g., S-nitrosylation) [810], or conformational changes due to substrate binding [11] result in the activation of the MMP.
Among the MMPs, the gelatinases (MMP-2 and MMP-9) as well as MMP-3 (stromelysin-1) and MMP-13 (collagenase-3) have been recognized as key players in the molecular mechanisms leading to neuronal injury in neurodegenerative conditions, traumatic brain injury, meningitis, multiple sclerosis, and stroke [4, 1215]. Damage to the blood–brain barrier (BBB) is a well-established neuropathological mechanism following brain injury, and activation of MMPs is critical in BBB breakdown under these conditions. Brain edema, hemorrhage, and neuronal death are the main consequences of BBB disruption following brain damage.
MMP-9 (gelatinase B) is significantly upregulated in animal models of cerebral ischemia [1621], traumatic brain injury [2226], and neuroinflammation [2729]. A large body of evidence strongly implicates MMP-9 in the detrimental molecular cascades leading to BBB dysfunction in these conditions. In stroke, MMP-9 plays a principal role in hemorrhagic conversion and vasogenic edema in animal models [16, 19, 21, 3034] and stroke patients [3540]. Severe neurological deficits and brain tissue loss positively correlate with elevated MMP-9 levels in stroke patients, who are at higher risk for developing life-threatening cerebral edema and hemorrhage [35, 37, 38, 4146].
Gelatin-substrate zymography is extensively used for the detection of gelatinase activity (MMP-2 and MMP-9) in biological samples [1]. This electrophoretic methodology is simple to perform and relatively sensitive, but may demand arduous work when a large number of samples need to be tested. A shortcoming of zymography is that it only provides a measure of total MMP-2/-9 expression (pro-MMPs plus active MMPs). Identification of active forms of these proteases in the gels is not always possible because of the low levels of expression, making net levels of activity for each gelatinase difficult to accurately quantify [47]. In addition, assessment of MMP-2/-9 expression in gelatin zymograms by quantifying active bands migrating at lower molecular weight might underestimate MMP activity since it has been shown that pro-MMPs could be activated by oxidative stress without proteolytic cleavage. This type of activation process does not appreciably change their molecular weight [9, 48].
The majority of MMP-9 is in a latent form, but it is the active form that exerts biological activity. Specific peptide substrates for MMP-9, and in general for any particular MMP, are very difficult to design because most MMPs share proteolytic recognition sequences [47, 4953]. Two methods have been described to specifically detect MMP activity in complex biological samples. Both use an immunocapture antibody to first immobilize the specific MMP to a 96-well plate followed by monitoring of the proteolytic activity using chromogenic or fluorogenic peptides [49, 54].
Based on these previously reported methods, here we describe the development of a high-throughput and highly sensitive MMP-9 activity assay able to detect endogenously active MMP-9 in biological samples. One of the main modifications in our assay is the initial coating of the plate with protein A/G and the use of a highly sensitive fluorescence resonance energy transfer (FRET) peptide substrate for MMP-9. By using protein A/G, which binds to the Fc region of immunoglobulins, we are able to immobilize and correctly orient the antibody for maximum MMP-9 immunocapture. This critical step in our methodology results in a more efficient and correct binding of the antibody to the plate compared to immobilization by passive absorption. The use of a commercially available FRET peptide having 5-carboxy-fluorescein (5-FAM) as a donor is another important improvement in our assay yielding a very strong fluorescent signal upon cleavage of the peptide by MMP-9. The use of other FRET pairs for measuring MMP activity have been proven to yield low fluorescence signals and consequently less sensitivity compared to 5-FAM peptides [55].
The general principle of our assay is presented in Figure 1. We validated this novel assay in complex biological matrices including rat brain homogenates, plasma, cell culture media from HT-1080 cancer cells, and RBE4 endothelial cell lysates. This sensitive fluorometric immunocapture assay specific for MMP-9 could be widely utilized to quantify active MMP-9 in biological samples and should be considered as a replacement for gelatin-based zymography. By changing the immunocapture antibody and FRET peptide, the activity of other members of the MMP family could be measured.

Results and discussion

Rat and human MMP-9 specifically and efficiently binds to mouse monoclonal anti-MMP-9

We first needed to determine the efficacy of the immunocapture antibody. The dose response of the mouse monoclonal anti-MMP-9 to rat and human recombinant MMP-9 (0.5, 1.0, 2.5, 5.0, and 10 ng) was examined (Figure 2A and 2B). Both rat and human MMP-9 fluorescent activity (measured in relative fluorescent units, or RFUs) increased as the quantity of recombinant MMP-9 increased. Figures 2C and 2D demonstrate the activity of “free” (no antibody coating on the plate, MMP-9 is added directly to the plate) versus immunocaptured rat and human MMP-9 (5 ng). No significant binding interference with the activity or activation of the gelatinase was observed with rat or human recombinant MMP-9. These findings indicate that binding of MMP-9 to the antibody is highly efficient as binding does not significantly reduce the catalytic ability of MMP-9.
A significant advantage in our methodology is to coat the black 96-well plate with protein G or use commercially available protein G coated plates. A more efficient immobilization of the anti-MMP-9 antibody to the plate results in a better sensitivity as shown in the Additional file 1: Figure S1. We compared the RFUs values for three different concentrations of active human recombinant MMP-9 between plates that were initially coated with protein G and non-coated plates. In the non-coated plates, the anti-MMP-9 antibody was immobilized by passive absorption to Fluotrac 600 high-binding plates overnight at 4°C. As presented in the Additional file 1: Figure S1, MMP-9 activity was significantly higher (p < 0.01) when the immunocapture antibody was immobilized to protein G-coated plates compared to plates in which the antibody was immobilized by passive absorption.
To study the specificity of the antibody for MMP-9, we assessed cross-reactivity with MMP-2. Because MMP-9 and MMP-2 have structural similarities, it was possible that MMP-2 binding and activation could contribute to the observed fluorescence. When recombinant rat MMP-2 was immunocaptured with the MMP-9 antibody, no detectable activity was observed (Figure 2E). This indicates rat MMP-2 is not recognized and captured by the MMP-9 antibody and is likely removed during a washing step. Recombinant human MMP-2 was immunocaptured at barely detectable activity levels, which indicates that human MMP-2, at very high doses, either will bind to the anti-MMP-9 or can bind to the protein G on the plate. However, the binding of human MMP-2 is probably not competitive with MMP-9 binding in biological samples. Rat and human MMP-2, pre-activated with APMA, was added to a plate with no antibody and combined with the FRET peptide. The free-floating MMP-2 (5 and 10 ng) was then able to interact with the FRET peptide to produce fluorescence (Figure 2E and 2F). This indicates that the lack of fluorescence when MMP-2 was immunocaptured with the mouse anti-MMP-9 is not due to lack of activity of the recombinant MMP-2. Specificity of the MMP-9 antibody for rat and human MMP-9 but not for rat or human MMP-2 was further demonstrated via immunoblotting (Figure 2G). Rat and human MMP-9 and MMP-2 were loaded in the gels. Initially, only MMP-9 was probed for using the mouse anti-MMP-9 antibody (shown in green). No bands were detected in the MMP-2 lanes, indicating that monoclonal MMP-9 antibody does not recognize rat or human MMP-2. To confirm the presence of MMP-2 in the gels, a second probe using an anti-MMP-2 antibody was performed (shown in red). These data indicate the monoclonal mouse antibody is highly specific for MMP-9.

Total and endogenously active levels of MMP-9 are measurable with the FRET peptide-based immunocapture assay in multiple biological samples

For this assay to be a viable technology in many research laboratories, it must be able to detect MMP-9 levels in many biological samples. We first demonstrated the ability of the assay to detect both total and endogenously active levels of MMP-9 in homogenates of rat cerebral cortex and plasma obtained from animals subjected to transient focal cerebral ischemia by middle cerebral artery occlusion (MCAO). Total and endogenously active levels of MMP-9 were recorded in ipsilateral (same hemisphere as ischemia) and contralateral (opposite hemisphere from ischemia) samples with 500 μg total protein (Figure 3A). Ipsilateral samples had more activity compared to contralateral samples for both total and active MMP-9. The addition of a classic matrix metalloproteinase inhibitor, GM6001 (total concentration 10 μM), to ipsilateral samples greatly reduced the activity of MMP-9. We have successfully detected total and endogenously active levels of MMP-9 in rat brain homogenates using as low as 50 μg total protein which emphasizes the high sensitivity of the assay (data not shown).
Figure 3B shows MMP-9 activity in 50 μL of rat plasma samples from sham-operated rats (surgery but no occlusion of the middle cerebral artery) and MCAO rats. Plasma from the sham rats had significantly less total MMP-9 activity than MCAO rats. Again, with the addition of GM6001 (10 μM) to the MCAO rat plasma, there was a large reduction in the activity of MMP-9. We also used the gelatinase inhibitor SB-3CT (0.5 μM and 5.0 μM) to check the specificity of the inhibition of MMP-9 in the plasma samples, as SB-3CT has very high selectivity to MMP-2 and MMP-9 compared to GM6001 [56, 57]. When SB-3CT was added to the MCAO plasma samples, there was a significant decrease in MMP-9 activity dependent on the concentration of SB-3CT added (Figure 3C).
This assay can also detect levels of human MMP-9 in conditioned cell culture media. As Figure 3D illustrates, increasing amounts of conditioned media (10, 50, and 100 μL) from the human fibrosarcoma cell line HT-1080, when immunocaptured with the mouse anti-MMP-9, coincides with greater MMP-9 activity.
Rat brain endothelial 4 (RBE4) cells are known to respond to inflammatory stimuli such as IL-1β [58]. We tested whether levels of MMP-9 can also be ascertained in cell lysates. Lysates were taken from RBE4 cells that were subjected to 24 h of stimulation with interleukin-1β (IL-1β; 10 ng/mL). When compared to control cell lysates, levels of MMP-9 in lysates from treated cells were significantly increased as seen by gelatin-substrate zymography (Figure 3E and 3F). When measured with the new FRET peptide assay, IL-1β-treated cell lysates had significantly more MMP-9 activity than controls (Figure 3G), indicating that this new assay demonstrates a similar trend in the MMP-9 levels in RBE4 cells after treatment with IL-1β as zymography.
Following ischemia/reperfusion, levels of MMP-9 are dramatically increased in the brain, specifically the ipsilateral hemisphere [32, 40]. Our FRET peptide-based assay reflects this well-known biological response and is sensitive enough to measure only endogenously active MMP-9 in samples, unlike zymography which primarily measures total levels of MMP-9. MMP-9 that is activated by oxidative stress is more easily quantified in this assay method as well, since activating pro-MMP-9 in this way does not substantially alter the molecular weight of MMP-9, therefore making it difficult to distinguish from the pro-MMP-9 in a zymogram. Inhibition of MMP-9 proteolytic activity by adding GM6001 to brain tissue, and GM6001 as well as SB-3CT in rat plasma, greatly reduced the emitted fluorescence, indicating it is MMP-9 cleaving the FRET peptide and not a different protease. That this assay also works in cultured cell media and cell lysates indicates it is not limited in sample medium as it can assess MMP-9 levels in multiple biological samples.

MMP-9 activity correlates in gelatin-substrate zymography and FRET peptide-based immunocapture assay

Gelatin-substrate zymography is a popular and accepted method for measuring levels of MMP-2/-9 in samples. However, if large amounts of samples need to be tested, repeated zymography can be very time-consuming. For all of this effort, only total levels of MMP-2/-9 can be accurately detected rather than both active and total levels of the gelatinases. Our proposed FRET peptide-based immunocapture assay provides the ability for many samples to be tested at once while also allowing for a choice of total or endogenously active levels of MMP-9 to be quantified. For this assay to be a viable replacement of gelatin-substrate zymography, relative fluorescence units (RFUs) observed for a sample in the immunocapture assay must be similar to relative lysis units (RLUs) recorded from gelatin zymography. To check for this correlation, total MMP-9 levels were studied from 16 rat MCAO cerebral cortex homogenates (ipsilateral and contralateral) in both the immunocapture assay and gelatin-substrate zymography. A sample zymogram of rat cortex samples and HT-1080 conditioned media indicating both gelatinases is shown in Figure 4A. As seen in the FRET peptide assay (Figure 3A), the zymogram data illustrates MMP-9 activity in ipsilateral samples as greater than contralateral samples. To determine whether the data from each method are comparable, we plotted the RLUs from the zymograms against the RFUs obtained from the FRET assay (Figure 4B). Total MMP-9 activity levels highly correlate between the two methods, indicating MMP-9 activity reported from zymography is very similar to that determined from the FRET peptide-based assay. These data indicate that the FRET-based immunocapture assay is a possible quantitative replacement of gelatin zymography for measurement of MMP-9.
Another advantage of the FRET-based assay is the flexibility offered. Other MMPs can easily be measured by altering the specific immunocapture antibody and/or the FRET peptide used. While gelatin zymography is limited to measurement of only total MMP-2 and MMP-9 activity, the FRET peptide assay can quantify total and active levels of the gelatinases.

Conclusion

Gelatin-substrate zymography is the go-to method for measurement of gelatinase activity in samples despite lengthy experiment times and restrictions on observable data. A new method, the FRET peptide-based immunocapture assay, has increased throughput, flexibility, and sensitivity and is proposed here as a viable quantitative replacement of zymography.

Materials and methods

Materials

The protein G coated 96-well plates and the mouse monoclonal anti-MMP-9 antibody were purchased from Thermo Fisher Scientific (Cat. Nos. 15157 and MS-817-P, respectively; Rockford, IL, USA). The FRET peptide was purchased from AnaSpec (Cat. No. 60570–01; San Jose, CA, USA). 4-aminophenylmercuric acetate (APMA), GM6001, and SB-3CT were purchased from Sigma-Aldrich (Cat. Nos. A9563-5G, M5939, and S1326, respectively; St. Louis, MO, USA).
Rat and human recombinant MMP-9 were purchased from R&D Systems (Cat. Nos. 5427-MM and 911-MP-010; Minneapolis, MN, USA). Rat and human recombinant MMP-2 were purchased from Abcam (Cat. Nos. ab39304 and ab40964; Cambridge, MA, USA). Crude brain tissue homogenates were prepared from rats with right side focal ischemia. Unfixed frozen cortex samples from both the ipsilateral and contralateral hemispheres were weighed and homogenized in 1% sodium dodecyl sulfate (SDS) buffer containing 150 mM NaCl, 50 mM Tris–HCl pH 7.6, 1% IGEPAL® CA-630, and 1% sodium deoxycholate. Just before the buffer was added to the brain samples, HALT Protease Inhibitor Cocktail (Cat. No. 78430; Thermo Fisher Scientific), HALT Phosphatase Inhibitor cocktail (Cat. No. 78428; Thermo Fisher Scientific) and 0.5 M EDTA were added at 10 μL/mL of homogenization buffer. Chilled tissue was homogenized in the buffer for 15-20 s using a Tissue-Tearor homogenizer (Cat. No. 985370; BioSpec, Inc, Bartlesville, OK, USA) then sonicated twice using a Vibra-Cell™ sonicator (Model VCX130PB; Sonics & Materials, Inc, Newtown, CT, USA). Resulting tissue homogenates were centrifuged at 14,000 xg for 20 min at 4°C in an Eppendorf microcentrifuge Model 5430R, and the supernatants aliquoted and stored at −80°C until used.

Rat stroke model and sample preparation

Focal cerebral ischemia was induced by temporary middle cerebral artery occlusion (MCAO) in male Wistar rats (280–320 g; Harlan Laboratories, Indianapolis, IN, USA) using the intraluminal filament method as described previously by our group [32, 59]. Briefly, rats were anesthetized with isoflurane in medical-grade oxygen and a midline vertical incision was made in the neck to expose the common carotid artery (CCA), external carotid artery (ECA) and internal carotid artery (ICA). The CCA was ligated permanently with a 4–0 silk suture and a vascular clip was temporarily placed in the pterygopalatine artery to prevent incorrect insertion of the occluding filament. A loose tie was placed over the ICA and ECA bifurcation with 4–0 silk suture and vascular clips were placed in the ICA and ECA. A small arteriotomy was made in the CCA approximately 2 mm proximal to the carotid bifurcation. A 4–0 silicone-coated filament (Cat. No. 403523PK10; Doccol Corporation, Sharon, MA, USA) was inserted through the CCA and advanced 18–20 mm inside the ICA until a mild resistance was felt. The occluding filament was left in place for 90 min and animals were allowed to recover from anesthesia. Eight to ten minutes before the end of the occlusion period, animals were re-anesthetized with isoflurane inhalant anesthesia, and the filament was gently retracted to allow reperfusion of the MCA territory.
After 48 h of reperfusion, animals were deeply anesthetized with pentobarbital (150 mg/kg; i.p.) and a blood sample was withdrawn from the vena cava into a heparinized syringe. Blood (1.5 mL) was quickly mixed with 50 μL of heparin (1000 U/mL) and centrifuged for 10 min at 2,000 xg to obtain the plasma. Rats were perfused intracardially with ice-cold saline and brains were harvested and dissected into ipsilateral (stroke side) and contralateral cerebral cortex and striatum. Samples were immediately frozen on dry ice and stored at −80°C until use.

Cell culture

HT-1080 human fibrosarcoma cells were obtained from American Type Culture Collection (ATCC, Manassas, VA, USA) and maintained in DMEM:F12 medium (Life Technologies, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS; Cat. No. 10082–147; Life Technologies), 100 U/mL penicillin and 100 μg/mL streptomycin in a humidified incubator at 37°C and 5% CO2. At 80-85% confluency, cells were washed with Dulbecco’s PBS and fresh media without FBS was added. After 24 h, cell culture media was collected and spun down at 5,000 xg for 10 min at 4°C. Aliquots of the HT-1080 conditioned media were prepared and stored at −80°C until use.
Rat brain endothelial (RBE4) cells were cultured in alpha-MEM/Ham’s F-10 Nutrient (1:1 solution; Cat. Nos. 12571–063 and 11550–043; GIBCO, Life Technologies) supplemented with 10% heat-inactivated fetal bovine serum (Cat. No. F4135; Sigma), 1% penicillin/streptomycin (Cat. No. 15140–122; GIBCO, Life Technologies), and 1% Geneticin (300 μg/mL; Cat. No. ALX-380-013-G001; Enzo Life Sciences). RBE4 cells were seeded in rat tail collagen I (50 μg/mL; Cat. No. C3867; Sigma) coated 6-well plates (20,000-30,000 cells/cm2) and maintained at 37°C, 5% CO2 incubator for 2 days before treatment. When cells reached 80-90% confluency, IL-1β (10 ng/mL; Cat. No. 501-RL/CF; R&D Systems, Inc., Minneapolis, MN, USA) was added to wells as treated groups. After 24 hours incubation, untreated and treated cells were washed once with ice-cold phosphate-buffered saline (PBS), and then lysed in radioimmunoprecipitation (RIPA) buffer consisting of 50 mM Tris–HCl (pH 7.6), 150 mM NaCl, 1% NP-40, 1% Sodium deoxycholate and 1% SDS plus complete protease and phosphatase inhibitor cocktails (Cat. No. 78430 and 78428; Thermo Scientific). Finally, the lysates were spun down at 14,000 xg for 15 min at 4°C. Aliquots of the supernatants were saved and stored at −80°C until use.

Immunocapture assay and fluorometric measurement of MMP-9 enzymatic activity

The activity of MMP-9 in rat stroke brain samples was measured fluorometrically using 5-FAM/QXL™520 FRET peptide (Cat. No. 60570; AnaSpec, San Jose, CA). In the intact FRET peptide (QXL™520-Pro-Leu-Gly-Cys[Me]-His-Ala-D-Arg-Lys[5-FAM]-NH2), the fluorescence of 5-FAM (5-carboxy-fluorescein) is quenched by the proximity of QXL™520. Upon cleavage into two fragments by MMP-9, the fluorescence of 5-FAM can be read and monitored at excitation/emission wavelengths of 485/528 nm.
Samples were prepared in 100 μL of TCNB buffer (50 mM Tris, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij® L23). Pierce protein G coated 96-well plates were washed three times with 200 μL TCNB buffer. Mouse monoclonal anti-MMP-9 (1 μg) in 100 μL of TCNB buffer was added to the wells and incubated for two hours at room temperature in a microplate mixer (USA Scientific, Ocala, FL, USA). Plates were then washed three times with 200 μL TCNB buffer and 100 μL of the samples were added to the wells in duplicate and allowed to incubate overnight at 4°C in a microplate mixer. After washing the plates three times with TCNB buffer, 1 mM APMA in 100 μL assay buffer (50 mM Tris–HCl pH 7.6, 200 mM NaCl, 5 mM CaCl2, 20 μM ZnCl, and 0.05% Brij® L23) was added to the wells to activate pro-MMP-9 in the samples to detect total levels of MMP-9, then covered in aluminum foil and incubated for 90 min at 37°C. To detect only endogenously active levels of MMP-9, APMA was not added and only 100 μL assay buffer was added to the wells. Next, 100 μL of assay buffer with 2 μM FRET peptide was added to the wells to bring total FRET peptide concentration to 1 μM. Fluorescence (relative fluorescence units, RFUs) was measured after 24 h of incubation at 37°C at 485 nm excitation and 528 nm emission in a Synergy HT Multi-mode microplate fluorescence reader (BioTek, Winooski, VT, USA) running Gen5™ data analysis software. A substrate control well was used to subtract baseline fluorescence from the sample wells.

Fluorometric MMP-2/-9 activity measurement without immunocapture

Recombinant MMPs were activated by adding APMA for a final concentration of 1 mM and incubated for 90 min at 37°C. Active MMPs were prepared in 100 μL assay buffer and mixed with 2 μM FRET peptide in 100 μL assay buffer for a final FRET peptide concentration of 1 μM. Fluorescence was monitored after a 24 h incubation period at 37°C at the same wavelengths mentioned above.

Gelatin-substrate zymography

Substrate-specific zymography for determination of activity of MMP-2 and MMP-9 was done on brain homogenates, HT-1080 conditioned media, and RBE4 cell lysates as described before [27, 60]. Brain homogenates or RBE4 cell lysates (50 μg total protein) were mixed in 10 μL zymogram sample buffer (Cat. No. 161–0764; Bio-Rad, Hercules, CA). 15 μL of HT-1080 conditioned media at 1:10 dilution in zymogram sample buffer was loaded in the gels. Proteins were separated by electrophoresis in a SDS-PAGE gel (8%) containing 0.1% (w/v) gelatin (Cat. No. G-2500; Sigma-Aldrich) at 150 V constant voltage. Gels were then washed twice in 2.5% Triton X-100 to remove SDS for 20 min then incubated for 24 h at 37°C in incubation buffer (50 mM Tris–HCl pH 7.6 containing 200 mM NaCl, 10 mM CaCl2, 0.02% Brij® L23, 0.02% NaN3). Gels were stained with Coomassie Brilliant Blue R solution (Cat. No. B6529; Sigma-Aldrich) for 1 h then destained in 10% acetic acid for one day before being scanned with an HP Scanjet 8300 scanner. Densitometric analysis of lytic zones at 94 and 88 kDa was performed using ImageJ (freely provided by NIH). The rat MMP-9 comprises two bands running in the zymography gels at 94 kDa (glycosylated form) and 88 kDa (intermediate form) [61].

Immunoblotting

Recombinant MMP-2 and MMP-9 were mixed in Laemmli sample buffer (Cat. No. 161–0737; Bio-Rad; Temecula, CA) containing 5% 2-mercaptoethanol and boiled for 10 min. Samples were loaded into TGX 4-20% precast gels (Cat. No. 456–1095; Bio-Rad) and subjected to electrophoresis. Proteins were transferred to Immobilon-FL polyvinylidene fluoride (PVDF) membranes using a Trans-Blot Turbo apparatus (Bio-Rad) at 25V for 30 min. Membranes were blocked for 1 h at room temperature with 5% non-fat milk in Tris-buffered saline (TBS). Membranes were incubated overnight at 4°C with primary antibodies against MMP-9 (mouse anti-MMP-9; Cat. No. MS-817-P; Thermo Scientific) and MMP-2 (rabbit anti-MMP-2; Cat. No. RPCA-MMP2; EnCor Biotechnology Inc., Gainesville, FL, USA) diluted 1:1000 in 5% milk in TBST. After washing with TBST (four times for 5 min each), membranes were incubated for 1 h at room temperature with goat anti-mouse IRDye 800CW secondary antibody (Cat. No. 926–32210; Li-Cor Biotechnology) diluted 1:30,000, and goat anti-rabbit 680RD antibody (Cat. No. 926–68071) diluted 1:40,000 in 5% milk in TBST. Protein bands were visualized using an Odyssey infrared scanner (Li-Cor Biosciences, Lincoln, NE, USA).

Statistical methods

GraphPad Prism 5 was used for statistical analysis. Statistical significance was assessed by ANOVA followed by Newman-Keuls corrections for multiple comparisons or with a Student’s t-test. Linear regression analysis was used to determine correlation information and significance. Bar graphs are shown as mean ± SEM. Probability p < 0.05 was considered statistically significant.

Acknowledgements

The authors would like to thank Dr. Michael Aschner at Vanderbilt University for kindly providing the RBE4 cell line. This work was supported by a research grant from the National Institute of Neurological Disorders and Stroke (NINDS; R01 grant NS065849) to ECJ. Additional support for this project was provided by the Brain and Spinal Cord Injury Research Trust Fund, McKnight Brain Institute, University of Florida.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KEH, GAR, and ECJ conceived and designed research; ECJ led the project; KEH, KMD, CY, and ECJ performed experiments; and KEH and ECJ performed data analysis and wrote the paper. All authors read and approved the final manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Birkedal-Hansen H, Yamada S, Windsor J, Pollard AH, Lyons G, Stetler-Stevenson W, Birkedal-Hansen B: Matrix metalloproteinases. Current protocols in cell biology. Edited by: Boni JS. 2008, Chapter 10:Unit 10 18 Birkedal-Hansen H, Yamada S, Windsor J, Pollard AH, Lyons G, Stetler-Stevenson W, Birkedal-Hansen B: Matrix metalloproteinases. Current protocols in cell biology. Edited by: Boni JS. 2008, Chapter 10:Unit 10 18
2.
Zurück zum Zitat Zuo J, Ferguson TA, Hernandez YJ, Stetler-Stevenson WG, Muir D: Neuronal matrix metalloproteinase-2 degrades and inactivates a neurite-inhibiting chondroitin sulfate proteoglycan. J Neurosci. 1998, 18 (14): 5203-5211.PubMed Zuo J, Ferguson TA, Hernandez YJ, Stetler-Stevenson WG, Muir D: Neuronal matrix metalloproteinase-2 degrades and inactivates a neurite-inhibiting chondroitin sulfate proteoglycan. J Neurosci. 1998, 18 (14): 5203-5211.PubMed
3.
Zurück zum Zitat Sternlicht MD, Werb Z: How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol. 2001, 17: 463-516. 10.1146/annurev.cellbio.17.1.463.PubMedCentralCrossRefPubMed Sternlicht MD, Werb Z: How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol. 2001, 17: 463-516. 10.1146/annurev.cellbio.17.1.463.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Candelario-Jalil E, Yang Y, Rosenberg GA: Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience. 2009, 158 (3): 983-994. 10.1016/j.neuroscience.2008.06.025.PubMedCentralCrossRefPubMed Candelario-Jalil E, Yang Y, Rosenberg GA: Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience. 2009, 158 (3): 983-994. 10.1016/j.neuroscience.2008.06.025.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat 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 (14): 5578-5582. 10.1073/pnas.87.14.5578.PubMedCentralCrossRefPubMed 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 (14): 5578-5582. 10.1073/pnas.87.14.5578.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Ogata Y, Enghild JJ, Nagase H: Matrix metalloproteinase 3 (stromelysin) activates the precursor for the human matrix metalloproteinase 9. J Biol Chem. 1992, 267 (6): 3581-3584.PubMed Ogata Y, Enghild JJ, Nagase H: Matrix metalloproteinase 3 (stromelysin) activates the precursor for the human matrix metalloproteinase 9. J Biol Chem. 1992, 267 (6): 3581-3584.PubMed
7.
Zurück zum Zitat O’Connell JP, Willenbrock F, Docherty AJ, Eaton D, Murphy G: Analysis of the role of the COOH-terminal domain in the activation, proteolytic activity, and tissue inhibitor of metalloproteinase interactions of gelatinase B. J Biol Chem. 1994, 269 (21): 14967-14973.PubMed O’Connell JP, Willenbrock F, Docherty AJ, Eaton D, Murphy G: Analysis of the role of the COOH-terminal domain in the activation, proteolytic activity, and tissue inhibitor of metalloproteinase interactions of gelatinase B. J Biol Chem. 1994, 269 (21): 14967-14973.PubMed
8.
Zurück zum Zitat Gu Z, Kaul M, Yan B, Kridel SJ, Cui J, Strongin A, Smith JW, Liddington RC, Lipton SA: S-nitrosylation of matrix metalloproteinases: signaling pathway to neuronal cell death. Science. 2002, 297 (5584): 1186-1190. 10.1126/science.1073634.CrossRefPubMed Gu Z, Kaul M, Yan B, Kridel SJ, Cui J, Strongin A, Smith JW, Liddington RC, Lipton SA: S-nitrosylation of matrix metalloproteinases: signaling pathway to neuronal cell death. Science. 2002, 297 (5584): 1186-1190. 10.1126/science.1073634.CrossRefPubMed
9.
Zurück zum Zitat Okamoto T, Akaike T, Sawa T, Miyamoto Y, van der Vliet A, Maeda H: Activation of matrix metalloproteinases by peroxynitrite-induced protein S-glutathiolation via disulfide S-oxide formation. J Biol Chem. 2001, 276 (31): 29596-29602. 10.1074/jbc.M102417200.CrossRefPubMed Okamoto T, Akaike T, Sawa T, Miyamoto Y, van der Vliet A, Maeda H: Activation of matrix metalloproteinases by peroxynitrite-induced protein S-glutathiolation via disulfide S-oxide formation. J Biol Chem. 2001, 276 (31): 29596-29602. 10.1074/jbc.M102417200.CrossRefPubMed
10.
Zurück zum Zitat Fu X, Kassim SY, Parks WC, Heinecke JW: Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J Biol Chem. 2001, 276 (44): 41279-41287. 10.1074/jbc.M106958200.CrossRefPubMed Fu X, Kassim SY, Parks WC, Heinecke JW: Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J Biol Chem. 2001, 276 (44): 41279-41287. 10.1074/jbc.M106958200.CrossRefPubMed
11.
Zurück zum Zitat Bannikov GA, Karelina TV, Collier IE, Marmer BL, Goldberg GI: Substrate binding of gelatinase B induces its enzymatic activity in the presence of intact propeptide. J Biol Chem. 2002, 277 (18): 16022-16027. 10.1074/jbc.M110931200.CrossRefPubMed Bannikov GA, Karelina TV, Collier IE, Marmer BL, Goldberg GI: Substrate binding of gelatinase B induces its enzymatic activity in the presence of intact propeptide. J Biol Chem. 2002, 277 (18): 16022-16027. 10.1074/jbc.M110931200.CrossRefPubMed
12.
Zurück zum Zitat Rosenberg GA: Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009, 8 (2): 205-216. 10.1016/S1474-4422(09)70016-X.CrossRefPubMed Rosenberg GA: Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009, 8 (2): 205-216. 10.1016/S1474-4422(09)70016-X.CrossRefPubMed
13.
Zurück zum Zitat Wang X, Rosell A, Lo EH: Targeting extracellular matrix proteolysis for hemorrhagic complications of tPA stroke therapy. CNS Neurol Disord Drug Targets. 2008, 7 (3): 235-242. 10.2174/187152708784936635.CrossRefPubMed Wang X, Rosell A, Lo EH: Targeting extracellular matrix proteolysis for hemorrhagic complications of tPA stroke therapy. CNS Neurol Disord Drug Targets. 2008, 7 (3): 235-242. 10.2174/187152708784936635.CrossRefPubMed
14.
Zurück zum Zitat Rosell A, Lo EH: Multiphasic roles for matrix metalloproteinases after stroke. Curr Opin Pharmacol. 2008, 8 (1): 82-89. 10.1016/j.coph.2007.12.001.CrossRefPubMed Rosell A, Lo EH: Multiphasic roles for matrix metalloproteinases after stroke. Curr Opin Pharmacol. 2008, 8 (1): 82-89. 10.1016/j.coph.2007.12.001.CrossRefPubMed
15.
Zurück zum Zitat Candelario-Jalil E, Thompson J, Taheri S, Grossetete M, Adair JC, Edmonds E, Prestopnik J, Wills J, Rosenberg GA: Matrix metalloproteinases are associated with increased blood–brain barrier opening in vascular cognitive impairment. Stroke. 2011, 42 (5): 1345-1350. 10.1161/STROKEAHA.110.600825.PubMedCentralCrossRefPubMed Candelario-Jalil E, Thompson J, Taheri S, Grossetete M, Adair JC, Edmonds E, Prestopnik J, Wills J, Rosenberg GA: Matrix metalloproteinases are associated with increased blood–brain barrier opening in vascular cognitive impairment. Stroke. 2011, 42 (5): 1345-1350. 10.1161/STROKEAHA.110.600825.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Romanic AM, White RF, Arleth AJ, Ohlstein EH, Barone FC: Matrix metalloproteinase expression increases after cerebral focal ischemia in rats: inhibition of matrix metalloproteinase-9 reduces infarct size. Stroke. 1998, 29 (5): 1020-1030. 10.1161/01.STR.29.5.1020.CrossRefPubMed Romanic AM, White RF, Arleth AJ, Ohlstein EH, Barone FC: Matrix metalloproteinase expression increases after cerebral focal ischemia in rats: inhibition of matrix metalloproteinase-9 reduces infarct size. Stroke. 1998, 29 (5): 1020-1030. 10.1161/01.STR.29.5.1020.CrossRefPubMed
17.
Zurück zum Zitat Rosenberg GA, Cunningham LA, Wallace J, Alexander S, Estrada EY, Grossetete M, Razhagi A, Miller K, Gearing A: Immunohistochemistry of matrix metalloproteinases in reperfusion injury to rat brain: activation of MMP-9 linked to stromelysin-1 and microglia in cell cultures. Brain Res. 2001, 893 (1–2): 104-112.CrossRefPubMed Rosenberg GA, Cunningham LA, Wallace J, Alexander S, Estrada EY, Grossetete M, Razhagi A, Miller K, Gearing A: Immunohistochemistry of matrix metalloproteinases in reperfusion injury to rat brain: activation of MMP-9 linked to stromelysin-1 and microglia in cell cultures. Brain Res. 2001, 893 (1–2): 104-112.CrossRefPubMed
18.
Zurück zum Zitat Rosenberg GA, Navratil M, Barone F, Feuerstein G: Proteolytic cascade enzymes increase in focal cerebral ischemia in rat. J Cereb Blood Flow Metab. 1996, 16 (3): 360-366.CrossRefPubMed Rosenberg GA, Navratil M, Barone F, Feuerstein G: Proteolytic cascade enzymes increase in focal cerebral ischemia in rat. J Cereb Blood Flow Metab. 1996, 16 (3): 360-366.CrossRefPubMed
19.
Zurück zum Zitat Park KP, Rosell A, Foerch C, Xing C, Kim WJ, Lee S, Opdenakker G, Furie KL, Lo EH: Plasma and brain matrix metalloproteinase-9 after acute focal cerebral ischemia in rats. Stroke. 2009, 40 (8): 2836-2842. 10.1161/STROKEAHA.109.554824.PubMedCentralCrossRefPubMed Park KP, Rosell A, Foerch C, Xing C, Kim WJ, Lee S, Opdenakker G, Furie KL, Lo EH: Plasma and brain matrix metalloproteinase-9 after acute focal cerebral ischemia in rats. Stroke. 2009, 40 (8): 2836-2842. 10.1161/STROKEAHA.109.554824.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Heo JH, Lucero J, Abumiya T, Koziol JA, Copeland BR, del Zoppo GJ: Matrix metalloproteinases increase very early during experimental focal cerebral ischemia. J Cereb Blood Flow Metab. 1999, 19 (6): 624-633.CrossRefPubMed Heo JH, Lucero J, Abumiya T, Koziol JA, Copeland BR, del Zoppo GJ: Matrix metalloproteinases increase very early during experimental focal cerebral ischemia. J Cereb Blood Flow Metab. 1999, 19 (6): 624-633.CrossRefPubMed
21.
Zurück zum Zitat Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH: Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab. 2000, 20 (12): 1681-1689.CrossRefPubMed Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH: Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab. 2000, 20 (12): 1681-1689.CrossRefPubMed
22.
Zurück zum Zitat Tejima E, Guo S, Murata Y, Arai K, Lok J, van Leyen K, Rosell A, Wang X, Lo EH: Neuroprotective effects of overexpressing tissue inhibitor of metalloproteinase TIMP-1. J Neurotrauma. 2009, 26 (11): 1935-1941. 10.1089/neu.2009.0959.PubMedCentralCrossRefPubMed Tejima E, Guo S, Murata Y, Arai K, Lok J, van Leyen K, Rosell A, Wang X, Lo EH: Neuroprotective effects of overexpressing tissue inhibitor of metalloproteinase TIMP-1. J Neurotrauma. 2009, 26 (11): 1935-1941. 10.1089/neu.2009.0959.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Shigemori Y, Katayama Y, Mori T, Maeda T, Kawamata T: Matrix metalloproteinase-9 is associated with blood–brain barrier opening and brain edema formation after cortical contusion in rats. Acta Neurochir Suppl. 2006, 96: 130-133. 10.1007/3-211-30714-1_29.CrossRefPubMed Shigemori Y, Katayama Y, Mori T, Maeda T, Kawamata T: Matrix metalloproteinase-9 is associated with blood–brain barrier opening and brain edema formation after cortical contusion in rats. Acta Neurochir Suppl. 2006, 96: 130-133. 10.1007/3-211-30714-1_29.CrossRefPubMed
24.
Zurück zum Zitat Higashida T, Kreipke CW, Rafols JA, Peng C, Schafer S, Schafer P, Ding JY, Dornbos D, Li X, Guthikonda M: The role of hypoxia-inducible factor-1alpha, aquaporin-4, and matrix metalloproteinase-9 in blood–brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg. 2011, 114 (1): 92-101. 10.3171/2010.6.JNS10207.CrossRefPubMed Higashida T, Kreipke CW, Rafols JA, Peng C, Schafer S, Schafer P, Ding JY, Dornbos D, Li X, Guthikonda M: The role of hypoxia-inducible factor-1alpha, aquaporin-4, and matrix metalloproteinase-9 in blood–brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg. 2011, 114 (1): 92-101. 10.3171/2010.6.JNS10207.CrossRefPubMed
25.
Zurück zum Zitat Wang X, Jung J, Asahi M, Chwang W, Russo L, Moskowitz MA, Dixon CE, Fini ME, Lo EH: Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci. 2000, 20 (18): 7037-7042.PubMed Wang X, Jung J, Asahi M, Chwang W, Russo L, Moskowitz MA, Dixon CE, Fini ME, Lo EH: Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci. 2000, 20 (18): 7037-7042.PubMed
26.
Zurück zum Zitat Mori T, Wang X, Aoki T, Lo EH: Downregulation of matrix metalloproteinase-9 and attenuation of edema via inhibition of ERK mitogen activated protein kinase in traumatic brain injury. J Neurotrauma. 2002, 19 (11): 1411-1419. 10.1089/089771502320914642.CrossRefPubMed Mori T, Wang X, Aoki T, Lo EH: Downregulation of matrix metalloproteinase-9 and attenuation of edema via inhibition of ERK mitogen activated protein kinase in traumatic brain injury. J Neurotrauma. 2002, 19 (11): 1411-1419. 10.1089/089771502320914642.CrossRefPubMed
27.
Zurück zum Zitat Candelario-Jalil E, Taheri S, Yang Y, Sood R, Grossetete M, Estrada EY, Fiebich BL, Rosenberg GA: Cyclooxygenase inhibition limits blood–brain barrier disruption following intracerebral injection of tumor necrosis factor-alpha in the rat. J Pharmacol Exp Ther. 2007, 323 (2): 488-498. 10.1124/jpet.107.127035.CrossRefPubMed Candelario-Jalil E, Taheri S, Yang Y, Sood R, Grossetete M, Estrada EY, Fiebich BL, Rosenberg GA: Cyclooxygenase inhibition limits blood–brain barrier disruption following intracerebral injection of tumor necrosis factor-alpha in the rat. J Pharmacol Exp Ther. 2007, 323 (2): 488-498. 10.1124/jpet.107.127035.CrossRefPubMed
28.
Zurück zum Zitat Meli DN, Christen S, Leib SL: Matrix metalloproteinase-9 in pneumococcal meningitis: activation via an oxidative pathway. J Infect Dis. 2003, 187 (9): 1411-1415. 10.1086/374644.CrossRefPubMed Meli DN, Christen S, Leib SL: Matrix metalloproteinase-9 in pneumococcal meningitis: activation via an oxidative pathway. J Infect Dis. 2003, 187 (9): 1411-1415. 10.1086/374644.CrossRefPubMed
29.
Zurück zum Zitat Leib SL, Clements JM, Lindberg RL, Heimgartner C, Loeffler JM, Pfister LA, Tauber MG, Leppert D: Inhibition of matrix metalloproteinases and tumour necrosis factor alpha converting enzyme as adjuvant therapy in pneumococcal meningitis. Brain. 2001, 124 (Pt 9): 1734-1742.CrossRefPubMed Leib SL, Clements JM, Lindberg RL, Heimgartner C, Loeffler JM, Pfister LA, Tauber MG, Leppert D: Inhibition of matrix metalloproteinases and tumour necrosis factor alpha converting enzyme as adjuvant therapy in pneumococcal meningitis. Brain. 2001, 124 (Pt 9): 1734-1742.CrossRefPubMed
30.
Zurück zum Zitat Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz MA, Fini ME, Lo EH: Effects of matrix metalloproteinase-9 gene knock-out on the proteolysis of blood–brain barrier and white matter components after cerebral ischemia. J Neurosci. 2001, 21 (19): 7724-7732.PubMed Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz MA, Fini ME, Lo EH: Effects of matrix metalloproteinase-9 gene knock-out on the proteolysis of blood–brain barrier and white matter components after cerebral ischemia. J Neurosci. 2001, 21 (19): 7724-7732.PubMed
31.
Zurück zum Zitat Gu Z, Cui J, Brown S, Fridman R, Mobashery S, Strongin AY, Lipton SA: A highly specific inhibitor of matrix metalloproteinase-9 rescues laminin from proteolysis and neurons from apoptosis in transient focal cerebral ischemia. J Neurosci. 2005, 25 (27): 6401-6408. 10.1523/JNEUROSCI.1563-05.2005.CrossRefPubMed Gu Z, Cui J, Brown S, Fridman R, Mobashery S, Strongin AY, Lipton SA: A highly specific inhibitor of matrix metalloproteinase-9 rescues laminin from proteolysis and neurons from apoptosis in transient focal cerebral ischemia. J Neurosci. 2005, 25 (27): 6401-6408. 10.1523/JNEUROSCI.1563-05.2005.CrossRefPubMed
32.
Zurück zum Zitat Rosenberg GA, Estrada EY, Dencoff JE: Matrix metalloproteinases and TIMPs are associated with blood–brain barrier opening after reperfusion in rat brain. Stroke. 1998, 29 (10): 2189-2195. 10.1161/01.STR.29.10.2189.CrossRefPubMed Rosenberg GA, Estrada EY, Dencoff JE: Matrix metalloproteinases and TIMPs are associated with blood–brain barrier opening after reperfusion in rat brain. Stroke. 1998, 29 (10): 2189-2195. 10.1161/01.STR.29.10.2189.CrossRefPubMed
33.
Zurück zum Zitat Pfefferkorn T, Rosenberg GA: Closure of the blood–brain barrier by matrix metalloproteinase inhibition reduces rtPA-mediated mortality in cerebral ischemia with delayed reperfusion. Stroke. 2003, 34 (8): 2025-2030. 10.1161/01.STR.0000083051.93319.28.CrossRefPubMed Pfefferkorn T, Rosenberg GA: Closure of the blood–brain barrier by matrix metalloproteinase inhibition reduces rtPA-mediated mortality in cerebral ischemia with delayed reperfusion. Stroke. 2003, 34 (8): 2025-2030. 10.1161/01.STR.0000083051.93319.28.CrossRefPubMed
34.
Zurück zum Zitat Tsuji K, Aoki T, Tejima E, Arai K, Lee SR, Atochin DN, Huang PL, Wang X, Montaner J, Lo EH: Tissue plasminogen activator promotes matrix metalloproteinase-9 upregulation after focal cerebral ischemia. Stroke. 2005, 36 (9): 1954-1959. 10.1161/01.STR.0000177517.01203.eb.CrossRefPubMed Tsuji K, Aoki T, Tejima E, Arai K, Lee SR, Atochin DN, Huang PL, Wang X, Montaner J, Lo EH: Tissue plasminogen activator promotes matrix metalloproteinase-9 upregulation after focal cerebral ischemia. Stroke. 2005, 36 (9): 1954-1959. 10.1161/01.STR.0000177517.01203.eb.CrossRefPubMed
35.
Zurück zum Zitat Montaner J, Alvarez-Sabin J, Molina C, Angles A, Abilleira S, Arenillas J, Gonzalez MA, Monasterio J: Matrix metalloproteinase expression after human cardioembolic stroke: temporal profile and relation to neurological impairment. Stroke. 2001, 32 (8): 1759-1766. 10.1161/01.STR.32.8.1759.CrossRefPubMed Montaner J, Alvarez-Sabin J, Molina C, Angles A, Abilleira S, Arenillas J, Gonzalez MA, Monasterio J: Matrix metalloproteinase expression after human cardioembolic stroke: temporal profile and relation to neurological impairment. Stroke. 2001, 32 (8): 1759-1766. 10.1161/01.STR.32.8.1759.CrossRefPubMed
36.
Zurück zum Zitat Montaner J, Alvarez-Sabin J, Molina CA, Angles A, Abilleira S, Arenillas J, Monasterio J: Matrix metalloproteinase expression is related to hemorrhagic transformation after cardioembolic stroke. Stroke. 2001, 32 (12): 2762-2767. 10.1161/hs1201.99512.CrossRefPubMed Montaner J, Alvarez-Sabin J, Molina CA, Angles A, Abilleira S, Arenillas J, Monasterio J: Matrix metalloproteinase expression is related to hemorrhagic transformation after cardioembolic stroke. Stroke. 2001, 32 (12): 2762-2767. 10.1161/hs1201.99512.CrossRefPubMed
37.
Zurück zum Zitat Montaner J, Molina CA, Monasterio J, Abilleira S, Arenillas JF, Ribo M, Quintana M, Alvarez-Sabin J: Matrix metalloproteinase-9 pretreatment level predicts intracranial hemorrhagic complications after thrombolysis in human stroke. Circulation. 2003, 107 (4): 598-603. 10.1161/01.CIR.0000046451.38849.90.CrossRefPubMed Montaner J, Molina CA, Monasterio J, Abilleira S, Arenillas JF, Ribo M, Quintana M, Alvarez-Sabin J: Matrix metalloproteinase-9 pretreatment level predicts intracranial hemorrhagic complications after thrombolysis in human stroke. Circulation. 2003, 107 (4): 598-603. 10.1161/01.CIR.0000046451.38849.90.CrossRefPubMed
38.
Zurück zum Zitat Montaner J, Rovira A, Molina CA, Arenillas JF, Ribo M, Chacon P, Monasterio J, Alvarez-Sabin J: Plasmatic level of neuroinflammatory markers predict the extent of diffusion-weighted image lesions in hyperacute stroke. J Cereb Blood Flow Metab. 2003, 23 (12): 1403-1407.CrossRefPubMed Montaner J, Rovira A, Molina CA, Arenillas JF, Ribo M, Chacon P, Monasterio J, Alvarez-Sabin J: Plasmatic level of neuroinflammatory markers predict the extent of diffusion-weighted image lesions in hyperacute stroke. J Cereb Blood Flow Metab. 2003, 23 (12): 1403-1407.CrossRefPubMed
39.
Zurück zum Zitat Rosell A, Alvarez-Sabin J, Arenillas JF, Rovira A, Delgado P, Fernandez-Cadenas I, Penalba A, Molina CA, Montaner J: A matrix metalloproteinase protein array reveals a strong relation between MMP-9 and MMP-13 with diffusion-weighted image lesion increase in human stroke. Stroke. 2005, 36 (7): 1415-1420. 10.1161/01.STR.0000170641.01047.cc.CrossRefPubMed Rosell A, Alvarez-Sabin J, Arenillas JF, Rovira A, Delgado P, Fernandez-Cadenas I, Penalba A, Molina CA, Montaner J: A matrix metalloproteinase protein array reveals a strong relation between MMP-9 and MMP-13 with diffusion-weighted image lesion increase in human stroke. Stroke. 2005, 36 (7): 1415-1420. 10.1161/01.STR.0000170641.01047.cc.CrossRefPubMed
40.
Zurück zum Zitat Rosell A, Cuadrado E, Ortega-Aznar A, Hernandez-Guillamon M, Lo EH, Montaner J: MMP-9-positive neutrophil infiltration is associated to blood–brain barrier breakdown and basal lamina type IV collagen degradation during hemorrhagic transformation after human ischemic stroke. Stroke. 2008, 39 (4): 1121-1126. 10.1161/STROKEAHA.107.500868.CrossRefPubMed Rosell A, Cuadrado E, Ortega-Aznar A, Hernandez-Guillamon M, Lo EH, Montaner J: MMP-9-positive neutrophil infiltration is associated to blood–brain barrier breakdown and basal lamina type IV collagen degradation during hemorrhagic transformation after human ischemic stroke. Stroke. 2008, 39 (4): 1121-1126. 10.1161/STROKEAHA.107.500868.CrossRefPubMed
41.
Zurück zum Zitat Vukasovic I, Tesija-Kuna A, Topic E, Supanc V, Demarin V, Petrovcic M: Matrix metalloproteinases and their inhibitors in different acute stroke subtypes. Clin Chem Lab Med CCLM/FESCC. 2006, 44 (4): 428-434. Vukasovic I, Tesija-Kuna A, Topic E, Supanc V, Demarin V, Petrovcic M: Matrix metalloproteinases and their inhibitors in different acute stroke subtypes. Clin Chem Lab Med CCLM/FESCC. 2006, 44 (4): 428-434.
42.
Zurück zum Zitat Ning M, Furie KL, Koroshetz WJ, Lee H, Barron M, Lederer M, Wang X, Zhu M, Sorensen AG, Lo EH: Association between tPA therapy and raised early matrix metalloproteinase-9 in acute stroke. Neurology. 2006, 66 (10): 1550-1555. 10.1212/01.wnl.0000216133.98416.b4.CrossRefPubMed Ning M, Furie KL, Koroshetz WJ, Lee H, Barron M, Lederer M, Wang X, Zhu M, Sorensen AG, Lo EH: Association between tPA therapy and raised early matrix metalloproteinase-9 in acute stroke. Neurology. 2006, 66 (10): 1550-1555. 10.1212/01.wnl.0000216133.98416.b4.CrossRefPubMed
43.
Zurück zum Zitat Castellanos M, Leira R, Serena J, Pumar JM, Lizasoain I, Castillo J, Davalos A: Plasma metalloproteinase-9 concentration predicts hemorrhagic transformation in acute ischemic stroke. Stroke. 2003, 34 (1): 40-46. 10.1161/01.STR.0000046764.57344.31.CrossRefPubMed Castellanos M, Leira R, Serena J, Pumar JM, Lizasoain I, Castillo J, Davalos A: Plasma metalloproteinase-9 concentration predicts hemorrhagic transformation in acute ischemic stroke. Stroke. 2003, 34 (1): 40-46. 10.1161/01.STR.0000046764.57344.31.CrossRefPubMed
44.
Zurück zum Zitat Castellanos M, Sobrino T, Millan M, Garcia M, Arenillas J, Nombela F, Brea D, Perez de la Ossa N, Serena J, Vivancos J: Serum cellular fibronectin and matrix metalloproteinase-9 as screening biomarkers for the prediction of parenchymal hematoma after thrombolytic therapy in acute ischemic stroke: a multicenter confirmatory study. Stroke. 2007, 38 (6): 1855-1859. 10.1161/STROKEAHA.106.481556.CrossRefPubMed Castellanos M, Sobrino T, Millan M, Garcia M, Arenillas J, Nombela F, Brea D, Perez de la Ossa N, Serena J, Vivancos J: Serum cellular fibronectin and matrix metalloproteinase-9 as screening biomarkers for the prediction of parenchymal hematoma after thrombolytic therapy in acute ischemic stroke: a multicenter confirmatory study. Stroke. 2007, 38 (6): 1855-1859. 10.1161/STROKEAHA.106.481556.CrossRefPubMed
45.
Zurück zum Zitat Serena J, Blanco M, Castellanos M, Silva Y, Vivancos J, Moro MA, Leira R, Lizasoain I, Castillo J, Davalos A: The prediction of malignant cerebral infarction by molecular brain barrier disruption markers. Stroke. 2005, 36 (9): 1921-1926. 10.1161/01.STR.0000177870.14967.94.CrossRefPubMed Serena J, Blanco M, Castellanos M, Silva Y, Vivancos J, Moro MA, Leira R, Lizasoain I, Castillo J, Davalos A: The prediction of malignant cerebral infarction by molecular brain barrier disruption markers. Stroke. 2005, 36 (9): 1921-1926. 10.1161/01.STR.0000177870.14967.94.CrossRefPubMed
46.
Zurück zum Zitat England TJ, Bath PM, Sare GM, Geeganage C, Moulin T, O’Neill D, Woimant F, Christensen H, De Deyn P, Leys D: Asymptomatic hemorrhagic transformation of infarction and its relationship with functional outcome and stroke subtype: assessment from the Tinzaparin in Acute Ischaemic Stroke Trial. Stroke. 2010, 41 (12): 2834-2839. 10.1161/STROKEAHA.109.573063.CrossRefPubMed England TJ, Bath PM, Sare GM, Geeganage C, Moulin T, O’Neill D, Woimant F, Christensen H, De Deyn P, Leys D: Asymptomatic hemorrhagic transformation of infarction and its relationship with functional outcome and stroke subtype: assessment from the Tinzaparin in Acute Ischaemic Stroke Trial. Stroke. 2010, 41 (12): 2834-2839. 10.1161/STROKEAHA.109.573063.CrossRefPubMed
47.
Zurück zum Zitat Quesada AR, Barbacid MM, Mira E, Fernandez-Resa P, Marquez G, Aracil M: Evaluation of fluorometric and zymographic methods as activity assays for stromelysins and gelatinases. Clin Exp Metastasis. 1997, 15 (1): 26-32.CrossRefPubMed Quesada AR, Barbacid MM, Mira E, Fernandez-Resa P, Marquez G, Aracil M: Evaluation of fluorometric and zymographic methods as activity assays for stromelysins and gelatinases. Clin Exp Metastasis. 1997, 15 (1): 26-32.CrossRefPubMed
48.
Zurück zum Zitat Viappiani S, Nicolescu AC, Holt A, Sawicki G, Crawford BD, Leon H, van Mulligen T, Schulz R: Activation and modulation of 72kDa matrix metalloproteinase-2 by peroxynitrite and glutathione. Biochem Pharmacol. 2009, 77 (5): 826-834. 10.1016/j.bcp.2008.11.004.CrossRefPubMed Viappiani S, Nicolescu AC, Holt A, Sawicki G, Crawford BD, Leon H, van Mulligen T, Schulz R: Activation and modulation of 72kDa matrix metalloproteinase-2 by peroxynitrite and glutathione. Biochem Pharmacol. 2009, 77 (5): 826-834. 10.1016/j.bcp.2008.11.004.CrossRefPubMed
49.
Zurück zum Zitat Lauer-Fields JL, Nagase H, Fields GB: Development of a solid-phase assay for analysis of matrix metalloproteinase activity. J Biomolecular Tech JBT. 2004, 15 (4): 305-316. Lauer-Fields JL, Nagase H, Fields GB: Development of a solid-phase assay for analysis of matrix metalloproteinase activity. J Biomolecular Tech JBT. 2004, 15 (4): 305-316.
50.
Zurück zum Zitat Nagase H, Fields CG, Fields GB: Design and characterization of a fluorogenic substrate selectively hydrolyzed by stromelysin 1 (matrix metalloproteinase-3). J Biol Chem. 1994, 269 (33): 20952-20957.PubMed Nagase H, Fields CG, Fields GB: Design and characterization of a fluorogenic substrate selectively hydrolyzed by stromelysin 1 (matrix metalloproteinase-3). J Biol Chem. 1994, 269 (33): 20952-20957.PubMed
51.
Zurück zum Zitat Nagase H, Fields GB: Human matrix metalloproteinase specificity studies using collagen sequence-based synthetic peptides. Biopolymers. 1996, 40 (4): 399-416. 10.1002/(SICI)1097-0282(1996)40:4<399::AID-BIP5>3.0.CO;2-R.CrossRefPubMed Nagase H, Fields GB: Human matrix metalloproteinase specificity studies using collagen sequence-based synthetic peptides. Biopolymers. 1996, 40 (4): 399-416. 10.1002/(SICI)1097-0282(1996)40:4<399::AID-BIP5>3.0.CO;2-R.CrossRefPubMed
52.
Zurück zum Zitat Lauer-Fields JL, Sritharan T, Stack MS, Nagase H, Fields GB: Selective hydrolysis of triple-helical substrates by matrix metalloproteinase-2 and −9. J Biol Chem. 2003, 278 (20): 18140-18145. 10.1074/jbc.M211330200.CrossRefPubMed Lauer-Fields JL, Sritharan T, Stack MS, Nagase H, Fields GB: Selective hydrolysis of triple-helical substrates by matrix metalloproteinase-2 and −9. J Biol Chem. 2003, 278 (20): 18140-18145. 10.1074/jbc.M211330200.CrossRefPubMed
53.
Zurück zum Zitat Verheijen JH, Nieuwenbroek NM, Beekman B, Hanemaaijer R, Verspaget HW, Ronday HK, Bakker AH: Modified proenzymes as artificial substrates for proteolytic enzymes: colorimetric assay of bacterial collagenase and matrix metalloproteinase activity using modified pro-urokinase. Biochem J. 1997, 323 (Pt 3): 603-609.PubMedCentralCrossRefPubMed Verheijen JH, Nieuwenbroek NM, Beekman B, Hanemaaijer R, Verspaget HW, Ronday HK, Bakker AH: Modified proenzymes as artificial substrates for proteolytic enzymes: colorimetric assay of bacterial collagenase and matrix metalloproteinase activity using modified pro-urokinase. Biochem J. 1997, 323 (Pt 3): 603-609.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Hanemaaijer R, Visser H, Konttinen YT, Koolwijk P, Verheijen JH: A novel and simple immunocapture assay for determination of gelatinase-B (MMP-9) activities in biological fluids: saliva from patients with Sjogren’s syndrome contain increased latent and active gelatinase-B levels. Matrix Biol J Int Soc Matrix Biol. 1998, 17 (8–9): 657-665.CrossRef Hanemaaijer R, Visser H, Konttinen YT, Koolwijk P, Verheijen JH: A novel and simple immunocapture assay for determination of gelatinase-B (MMP-9) activities in biological fluids: saliva from patients with Sjogren’s syndrome contain increased latent and active gelatinase-B levels. Matrix Biol J Int Soc Matrix Biol. 1998, 17 (8–9): 657-665.CrossRef
55.
Zurück zum Zitat Highly sensitive FRET substrates for the assay of MMPs.http://www.anaspec.com/servePdf.asp?f=c_publications24.pdf&t=publication Highly sensitive FRET substrates for the assay of MMPs.http://​www.​anaspec.​com/​servePdf.​asp?​f=​c_​publications24.​pdf&​t=​publication
56.
Zurück zum Zitat Fisher JF, Mobashery S: Recent advances in MMP inhibitor design. Cancer Metastasis Rev. 2006, 25 (1): 115-136. 10.1007/s10555-006-7894-9.CrossRefPubMed Fisher JF, Mobashery S: Recent advances in MMP inhibitor design. Cancer Metastasis Rev. 2006, 25 (1): 115-136. 10.1007/s10555-006-7894-9.CrossRefPubMed
57.
Zurück zum Zitat Lee M, Bernardo MM, Meroueh SO, Brown S, Fridman R, Mobashery S: Synthesis of chiral 2-(4-phenoxyphenylsulfonylmethyl)thiiranes as selective gelatinase inhibitors. Org Lett. 2005, 7 (20): 4463-4465. 10.1021/ol0517269.CrossRefPubMed Lee M, Bernardo MM, Meroueh SO, Brown S, Fridman R, Mobashery S: Synthesis of chiral 2-(4-phenoxyphenylsulfonylmethyl)thiiranes as selective gelatinase inhibitors. Org Lett. 2005, 7 (20): 4463-4465. 10.1021/ol0517269.CrossRefPubMed
58.
Zurück zum Zitat Lok J, Zhao S, Leung W, Seo JH, Navaratna D, Wang X, Whalen MJ, Lo EH: Neuregulin-1 effects on endothelial and blood–brain-barrier permeability after experimental injury. Trans Stroke Res. 2012, 3 (S1): S119-S124. 10.1007/s12975-012-0157-x.CrossRef Lok J, Zhao S, Leung W, Seo JH, Navaratna D, Wang X, Whalen MJ, Lo EH: Neuregulin-1 effects on endothelial and blood–brain-barrier permeability after experimental injury. Trans Stroke Res. 2012, 3 (S1): S119-S124. 10.1007/s12975-012-0157-x.CrossRef
59.
Zurück zum Zitat Candelario-Jalil E, Gonzalez-Falcon A, Garcia-Cabrera M, Leon OS, Fiebich BL: Post-ischaemic treatment with the cyclooxygenase-2 inhibitor nimesulide reduces blood–brain barrier disruption and leukocyte infiltration following transient focal cerebral ischaemia in rats. J Neurochem. 2007, 100 (4): 1108-1120. 10.1111/j.1471-4159.2006.04280.x.CrossRefPubMed Candelario-Jalil E, Gonzalez-Falcon A, Garcia-Cabrera M, Leon OS, Fiebich BL: Post-ischaemic treatment with the cyclooxygenase-2 inhibitor nimesulide reduces blood–brain barrier disruption and leukocyte infiltration following transient focal cerebral ischaemia in rats. J Neurochem. 2007, 100 (4): 1108-1120. 10.1111/j.1471-4159.2006.04280.x.CrossRefPubMed
60.
Zurück zum Zitat Zhang JW, Gottschall PE: Zymographic measurement of gelatinase activity in brain tissue after detergent extraction and affinity-support purification. J Neurosci Methods. 1997, 76 (1): 15-20. 10.1016/S0165-0270(97)00065-4.CrossRefPubMed Zhang JW, Gottschall PE: Zymographic measurement of gelatinase activity in brain tissue after detergent extraction and affinity-support purification. J Neurosci Methods. 1997, 76 (1): 15-20. 10.1016/S0165-0270(97)00065-4.CrossRefPubMed
61.
Zurück zum Zitat Zhang JW, Deb S, Gottschall PE: Regional and differential expression of gelatinases in rat brain after systemic kainic acid or bicuculline administration. Eur J Neurosci. 1998, 10 (11): 3358-3368. 10.1046/j.1460-9568.1998.00347.x.CrossRefPubMed Zhang JW, Deb S, Gottschall PE: Regional and differential expression of gelatinases in rat brain after systemic kainic acid or bicuculline administration. Eur J Neurosci. 1998, 10 (11): 3358-3368. 10.1046/j.1460-9568.1998.00347.x.CrossRefPubMed
Metadaten
Titel
Fluorometric immunocapture assay for the specific measurement of matrix metalloproteinase-9 activity in biological samples: application to brain and plasma from rats with ischemic stroke
verfasst von
Kimberly E Hawkins
Kelly M DeMars
Changjun Yang
Gary A Rosenberg
Eduardo Candelario-Jalil
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Molecular Brain / Ausgabe 1/2013
Elektronische ISSN: 1756-6606
DOI
https://doi.org/10.1186/1756-6606-6-14

Weitere Artikel der Ausgabe 1/2013

Molecular Brain 1/2013 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.