MMP 13 Human

Matrix Metalloproteinase-13 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Key Substrates Degraded by MMP-13

SubstratePhysiological ContextPathological Context
Collagen I, II, IIIBone remodeling, cartilage turnoverArthritis, tumor invasion
Collagen IV, IX, X, XIVBasement membrane integrityAngiogenesis, metastasis
Aggrecan, perlecanCartilage matrix stabilityOsteoarthritis, atherosclerosis
FibronectinCell adhesion, wound healingTumor microenvironment remodeling

Table 1: MMP-13 Substrate Specificity

SubstrateDegradation EfficiencySource
Collagen IIHigh (primary target)
Collagen IModerate
AggrecanModerate
FibronectinLow

Skeletal Development and Bone Homeostasis

  • Growth plate remodeling: MMP-13-deficient mice exhibit delayed endochondral ossification, enlarged hypertrophic zones, and vascularization defects in primary ossification centers .

  • Cartilage turnover: Human chondrocytes constitutively express MMP-13, but its extracellular levels are tightly regulated via LRP1-mediated endocytosis. This balance prevents excessive matrix degradation in healthy cartilage .

Table 2: MMP-13 in Skeletal Biology

ProcessMMP-13 FunctionExperimental Evidence
Growth plate maturationCleaves type II collagenMmp13−/− mice show prolonged hypertrophic zones
Endochondral ossificationFacilitates vascular invasionDelayed primary ossification centers in Mmp13−/− embryos
Cartilage homeostasisLimited ECM degradation via LRP1 bindingRAP blockade reveals constitutive MMP-13 secretion in human chondrocytes

Atherosclerosis and Cardiovascular Disease

MMP-13 contributes to plaque instability by degrading collagen in fibrous caps. In apoE−/− mice, selective MMP-13 inhibition (MMP13i-A) increased plaque collagen content and fibrous cap thickness, reducing rupture risk .

Cancer and Metastasis

MMP-13 overexpression correlates with aggressive tumor phenotypes:

  • Pan-cancer analysis: High MMP-13 levels predict poor prognosis in multiple cancers (e.g., lung, breast, ovarian) .

  • Mechanisms: Drives epithelial-to-mesenchymal transition (EMT) and extracellular matrix remodeling .

Arthritis and Cartilage Degradation

  • Osteoarthritis (OA): MMP-13 is upregulated in chondrocytes, degrading aggrecan and type II collagen .

  • Rheumatoid Arthritis (RA): Inhibitors reduce cartilage erosion in collagen-induced arthritis (CIA) models but show limited efficacy in inflammatory models like antigen-induced arthritis (AIA) .

Animal Model Findings

ModelInterventionOutcomeSource
SCID mouse (RA)MMP-13 inhibitor (60 mg/kg)75% reduction in cartilage invasion
apoE−/− miceMMP13i-A (40 mg/kg)407% increase in plaque collagen
Mmp13−/− miceNoneDelayed bone mineralization

Clinical Relevance of MMP-13 Inhibitors

  • Selectivity: MMP13i-A (non-hydroxamic acid inhibitor) shows nanomolar potency against MMP-13 without affecting MMP-1, -2, -7, -9, or -14 .

  • Toxicity: Unlike earlier MMP inhibitors, MMP13i-A avoids musculoskeletal side effects in preclinical studies .

ELISA and IHC Applications

MethodApplicationSensitivitySource
Human MMP-13 ELISA KitSerum/plasma quantification78 pg/ml → 5000 pg/ml range
ImmunohistochemistryTumor/cancer tissue analysisDetects pro- and active MMP-13

Table 3: MMP-13 Detection Methods

TechnologyAdvantagesLimitations
qPCR primer pairsHigh specificity for MMP13 mRNARequires RNA extraction/purification
Fluorescent probesReal-time activity monitoringLimited to preclinical models

Product Specs

Introduction

Matrix metalloproteinase-13 (MMP-13) is an enzyme belonging to the MMP extracellular protease family. These proteases have broad substrate specificities and contribute to both normal and diseased tissue proliferation. MMP-13 targets various molecules, including collagen, gelatin, entactin, pro-TNF-alpha, and chemokine SDF-11-4. Initially, MMP-13 exists in its inactive form as a glycosylated proenzyme with a molecular weight of 52-56 kDa. Upon cleavage, it transforms into the active MMP-13 form, weighing 22-46 kDa, which plays a crucial role in extracellular matrix remodeling. Due to its significant involvement in cell migration and metastasis, MMP-13 is a key target for inhibition studies.

Description
The MMP-13 protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 391 amino acids (specifically, amino acids 104-471), resulting in a molecular weight of 44.7 kDa. This MMP-13 variant includes a 23 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The MMP-13 solution, at a concentration of 0.5 mg/ml, is prepared in a buffer containing 20 mM Tris-HCl (pH 8.0), 10% glycerol, 0.15 M NaCl, and 1 mM DTT.
Stability
For optimal storage, keep the MMP-13 at 4°C if you plan to use the entire vial within 2-4 weeks. For longer-term storage, freezing at -20°C is recommended. Adding a carrier protein, such as 0.1% HSA or BSA, is suggested for extended storage durations. Importantly, avoid repeatedly freezing and thawing the MMP-13 solution.
Purity

The purity of MMP-13 is determined to be greater than 85% using SDS-PAGE analysis.

Synonyms

CLG3, MANDP1, Matrix metalloproteinase-13, MMP-13, MMP13.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSYNVFPRT LKWSKMNLTY RIVNYTPDMT HSEVEKAFKK AFKVWSDVTP LNFTRLHDGI ADIMISFGIK EHGDFYPFDG PSGLLAHAFP PGPNYGGDAH FDDDETWTSS SKGYNLFLVA AHEFGHSLGL DHSKDPGALM FPIYTYTGKS HFMLPDDDVQ GIQSLYGPGD EDPNPKHPKT PDKCDPSLSL DAITSLRGET MIFKDRFFWR LHPQQVDAEL FLTKSFWPEL PNRIDAAYEH PSHDLIFIFR GRKFWALNGY DILEGYPKKI SELGLPKEVK KISAAVHFED TGKTLLFSGN QVWRYDDTNH IMDKDYPRLI EEDFPGIGDK VDAVYEKNGY IYFFNGPIQF EYSIWSNRIV RVMPANSILW C.

Q&A

What is the basic structure of human MMP-13 and how does it differ from other matrix metalloproteinases?

Human MMP-13 (collagenase-3) is a zinc-dependent endopeptidase that belongs to the matrix metalloproteinase family. Unlike many other human MMPs that are widely distributed, MMP-13 shows a restrictive distribution pattern in normal tissues and selective expression in pathological conditions . The enzyme contains several domains common to MMPs including a propeptide domain, catalytic domain, hinge region, and hemopexin domain.

What distinguishes MMP-13 is its substrate specificity and regulatory mechanisms. Unlike other MMPs, the human MMP-13 gene is transcribed into several different transcripts which could potentially yield proteins with activities and functions different from those of the original MMP-13 . Its promoter contains specific binding sites involved in transcriptional regulation, including TATA box, AP-1, PEA-3, OSE-2, and a negative regulator called AGRE .

What are the primary substrates of human MMP-13 and what determines its substrate specificity?

MMP-13 demonstrates remarkable versatility in substrate utilization. It is highly active against type II collagen, which is critical in cartilage degradation, but it also cleaves numerous other substrates:

Substrate CategorySpecific Substrates
CollagensType II collagen (primary), Types I, III, IV, IX, X, XIV
ECM ComponentsAggrecan, fibronectin, tenascin, perlecan
Non-ECM ProteinsConnective tissue growth factor (CTGF), fibrinogen
Cell Surface MoleculesVarious membrane receptors and adhesion molecules

The substrate specificity of MMP-13 is determined by its unique structural features, particularly within the catalytic domain and hemopexin-like domain, which create specific binding pockets that recognize target substrates . This specificity makes MMP-13 particularly relevant in pathological conditions affecting tissues rich in type II collagen.

How is MMP-13 normally expressed in healthy human tissues?

In contrast to many human MMPs that are widely distributed, MMP-13 exhibits a restrictive expression pattern in normal tissues . Under physiological conditions, MMP-13 expression is primarily found in:

  • Developing bone during fetal development

  • Areas of bone remodeling in adults

  • Wound healing processes

  • Certain reproductive tissues during specific phases

This limited expression pattern suggests tight regulation of MMP-13 under normal conditions, which becomes dysregulated in pathological states such as arthritis and cancer . Understanding this baseline expression is critical for interpreting research findings related to MMP-13 in disease states.

What role does MMP-13 play in cerebral ischemia and stroke?

MMP-13 activation and nuclear translocation appears to be an early consequence of ischemic stimulus in both animal models and human stroke patients . Studies have revealed several key findings:

  • Active MMP-13 protein increases significantly (P<0.05) after just 90 minutes of cerebral ischemia in rat models .

  • Human infarct/periinfarct samples show higher levels of active MMP-13 (P<0.05) compared with contralateral brain regions .

  • MMP-13 colocalizes with nuclear DAPI staining in both humans and rats, suggesting nuclear translocation after ischemia .

  • Cellular distribution varies between species: while neurons are the main producers in both humans and rats, MMP-13 is also found in oligodendrocytes in rats and astrocytes in humans .

This early activation of MMP-13 may initiate a cascade of MMP activation, as MMP-13 can activate MMPs-2 and -9, which have been detected at later timepoints (6 and 24 hours respectively) after stroke . Nuclear localization of MMP-13 could potentially contribute to DNA fragmentation during the apoptotic cascade following ischemia .

How is MMP-13 involved in arthritis pathogenesis and what makes it a potential therapeutic target?

MMP-13 plays a pivotal role in arthritis pathogenesis due to its high activity against type II collagen, the primary collagen in articular cartilage . Key aspects of its involvement include:

  • Elevated expression in osteoarthritic and rheumatoid arthritic cartilage compared to normal tissue

  • Contribution to cartilage matrix degradation through cleavage of type II collagen and aggrecan

  • Involvement in the inflammatory cascade through processing of cytokines and chemokines

  • Activation of other MMPs in a proteolytic cascade that amplifies tissue damage

What is known about MMP-13 expression and function in human cancers?

MMP-13 is selectively expressed in various human cancers and contributes to tumor progression through multiple mechanisms . Key findings include:

Cancer TypeMMP-13 RoleResearch Observations
Breast CancerPromotes invasion and metastasisHigher expression correlates with poorer prognosis
Head and Neck CancerECM degradation and tumor cell migrationOften localized at the invasive front of tumors
MelanomaFacilitates tissue invasionExpression increases with malignant progression
Colorectal CancerPromotes angiogenesis and invasionAssociated with advanced disease stages

MMP-13 contributes to cancer progression through:

  • Degradation of extracellular matrix components, facilitating tumor invasion

  • Release of growth factors sequestered in the ECM

  • Processing of cell surface proteins that mediate cell-cell and cell-matrix interactions

  • Participation in angiogenic processes necessary for tumor growth

Understanding these roles has led to interest in MMP-13 as both a biomarker and therapeutic target in certain cancers .

What are the optimal methods for detecting MMP-13 activation in tissue samples?

The detection of active MMP-13 in tissue samples requires specific techniques to distinguish between latent and active forms. Based on the research methodologies described, several complementary approaches are recommended:

  • Western Blot Analysis:

    • Protein extraction using specific buffers containing protease inhibitors (1 mmol/L phenylmethylsulfonyl fluoride and 7 μg/mL aprotinin)

    • SDS-PAGE (12%) separation followed by transfer to PVDF membranes

    • Immunodetection using specific anti-MMP-13 antibodies (e.g., mouse anti-MMP-13 at 1:300 dilution)

    • Visualization with chemiluminescent reagents

  • In Situ Zymography:

    • Unfixed frozen tissue sections incubated with fluorescein-labeled substrates

    • Inclusion of broad-spectrum MMP inhibitor (1,10-phenanthroline, 10 mmol/L) controls to ensure specificity

    • Analysis using confocal microscopy

  • Immunohistochemistry:

    • Fixation protocols optimized to maintain antigen integrity

    • Double immunostaining with cell-type specific markers (NeuN for neurons, GFAP for astrocytes, etc.)

    • Nuclear counterstaining with DAPI to assess nuclear localization

    • Z-stack imaging for 3D reconstruction to confirm spatial colocalization

Each method offers different advantages: Western blot quantifies total active MMP-13, zymography detects functional enzyme activity, and immunohistochemistry provides spatial information about cellular localization .

How can researchers establish reliable in vitro models to study MMP-13 function in neural cells?

Based on the search results, an effective in vitro model for studying MMP-13 function in neural cells would include:

  • Primary Neuronal Culture Establishment:

    • Isolation of rat cortical neurons (or appropriate human cell lines)

    • Maintenance in balanced salt solution medium supplemented with glucose (5.5 mmol/L)

    • Verification of neuronal phenotype using markers such as MAP-2

  • Oxygen and Glucose Deprivation (OGD) Protocol:

    • Replace medium with glucose-free balanced salt solution

    • Incubate cells in an anaerobic chamber (95% N₂, 5% CO₂) at 37°C

    • Control conditions: standard incubator with glucose-supplemented medium

  • Analysis Methods:

    • Nuclear fraction isolation for specific assessment of nuclear MMP-13

    • Immunocytochemistry with anti-MMP-13 antibodies and nuclear counterstaining

    • Functional assays to measure MMP-13 activity

    • Assessment of neuronal integrity using MAP-2 staining to evaluate neurite extensions

This model effectively reproduces the nuclear translocation of MMP-13 observed in vivo following ischemia, making it suitable for mechanistic studies of MMP-13 activation and function in neural cells .

What techniques are most effective for quantifying MMP-13 transcriptional regulation?

For investigating MMP-13 transcriptional regulation, several complementary techniques should be employed:

  • Promoter Analysis and Reporter Assays:

    • Cloning of the MMP-13 promoter region containing regulatory elements (TATA box, AP-1, PEA-3, OSE-2, AGRE)

    • Construction of reporter constructs with sequential deletions or specific mutations

    • Transfection into relevant cell types and measurement of reporter activity under various conditions

  • Chromatin Immunoprecipitation (ChIP):

    • Crosslinking of protein-DNA complexes in intact cells

    • Immunoprecipitation with antibodies against specific transcription factors

    • Analysis of bound DNA fragments by PCR or sequencing

    • Particularly useful for studying binding of factors to OSE-2, AP-1, and PEA-3 sites in the MMP-13 promoter

  • RNA Analysis Methods:

    • RT-PCR for detecting specific MMP-13 transcript variants

    • RNA stability assays to assess post-transcriptional regulation

    • RNA-seq for comprehensive transcriptome analysis

  • DNA-Protein Interaction Assays:

    • Electrophoretic mobility shift assays (EMSA) for studying binding of nuclear factors to specific promoter elements

    • DNA pulldown assays followed by mass spectrometry to identify novel regulatory proteins

These approaches allow comprehensive characterization of the multiple levels of MMP-13 transcriptional control, including the newly identified negative regulator AGRE .

How is MMP-13 activation regulated in human tissues?

MMP-13 activation is controlled through a multi-layered regulatory system:

  • Zymogen Activation:

    • MMP-13 is synthesized as an inactive proenzyme (pro-MMP-13)

    • Activation requires proteolytic removal of the propeptide domain

    • Involves a proteolytic cascade including MMP-14 (MT1-MMP) and MMP-2

    • Various proteases can cleave the propeptide, including plasmin, cathepsins, and other MMPs

  • Tissue Inhibitors of Metalloproteinases (TIMPs):

    • TIMPs bind to the active site of MMPs and block substrate access

    • TIMP-1, TIMP-2, TIMP-3, and TIMP-4 have varying affinities for MMP-13

    • Balance between MMP-13 and its inhibitors determines net proteolytic activity

  • Compartmentalization:

    • MMP-13 activity is regulated through subcellular localization

    • Nuclear translocation observed after ischemic stimulus in neural cells

    • Cell surface binding can concentrate MMP-13 activity at specific extracellular sites

  • Post-translational Modifications:

    • Phosphorylation, glycosylation, and nitrosylation can affect enzyme activity

    • Oxidative modifications can lead to autoactivation under stress conditions

Understanding these regulatory mechanisms is crucial for developing strategies to modulate MMP-13 activity in pathological conditions .

What signaling pathways control MMP-13 expression in response to inflammatory stimuli?

MMP-13 expression is regulated by multiple signaling pathways in response to inflammatory stimuli:

  • MAPK Pathways:

    • ERK1/2, p38, and JNK cascades activate transcription factors like AP-1

    • AP-1 (composed of c-Fos and c-Jun) binds to the AP-1 site in MMP-13 promoter

    • Pathway inhibitors can selectively block inflammation-induced MMP-13 expression

  • NF-κB Pathway:

    • Activated by proinflammatory cytokines like IL-1β and TNF-α

    • Regulates MMP-13 directly and through secondary mediators

    • Cross-talks with MAPK pathways to amplify inflammatory responses

  • JAK/STAT Pathway:

    • Mediates effects of cytokines like IL-6

    • STAT proteins bind to specific elements in the MMP-13 promoter

    • Important in stress responses following ischemia

  • Wnt/β-catenin Signaling:

    • Regulates MMP-13 expression in skeletal tissues

    • Interacts with OSE-2 binding elements in the MMP-13 promoter

    • Critical in pathological conditions like osteoarthritis

These pathways present potential targets for selective modulation of MMP-13 expression in inflammatory conditions, with implications for therapeutic development.

What is the relationship between mechanical stress and MMP-13 regulation in human tissues?

Mechanical stress is a significant regulator of MMP-13 expression, particularly in load-bearing tissues:

  • Mechanotransduction Pathways:

    • Mechanical stimuli activate integrin-mediated signaling

    • Cytoskeletal reorganization triggers kinase cascades (FAK, Src, MAPK)

    • These pathways converge on transcription factors that regulate MMP-13

  • Effects in Articular Cartilage:

    • Physiological loading suppresses MMP-13 expression in healthy chondrocytes

    • Abnormal mechanical stress (excessive or insufficient) induces MMP-13 upregulation

    • This mechanosensitive regulation is altered in osteoarthritis

  • Cellular Responses to Fluid Shear Stress:

    • Endothelial cells modulate MMP-13 expression in response to shear stress

    • Relevant in vascular remodeling and atherosclerotic plaque stability

    • May contribute to stroke pathophysiology through vascular matrix remodeling

  • Mechanically-Responsive Promoter Elements:

    • Specific elements in the MMP-13 promoter respond to mechanical stimuli

    • Includes response elements for AP-1 and PEA-3 transcription factors

    • Mechanical stimulation can override chemical regulatory mechanisms

Understanding these mechanosensitive regulatory mechanisms has implications for conditions where altered mechanical loading contributes to pathology, such as osteoarthritis, tendinopathies, and vascular diseases .

What is the significance of nuclear localization of MMP-13 in neural cells after ischemia?

The nuclear localization of MMP-13 in neural cells following ischemia represents a novel finding with potentially significant implications for stroke pathophysiology:

  • Mechanism of Nuclear Translocation:

    • MMP-13 contains putative nuclear localization signals (NLS)

    • Nuclear import may involve karyopherins/importins recognizing these signals

    • Translocation occurs rapidly (within 30 minutes) after ischemic stimulus

    • Process can be reproduced in vitro using oxygen-glucose deprivation models

  • Potential Nuclear Functions:

    • DNA fragmentation: MMP-13 may contribute to apoptotic DNA degradation

    • PARP cleavage: May cleave poly-(ADP-ribose) polymerase, contributing to cell death

    • Transcriptional regulation: Potential modification of nuclear proteins affecting gene expression

    • Chromatin remodeling: May process histones or other chromatin-associated proteins

  • Comparative Analysis of Cell Types:

    • Primary localization in neurons across species (human and rat)

    • Additional presence in rat oligodendrocytes but human astrocytes

    • These differences suggest species-specific roles in neural injury

  • Temporal Profile:

    • Nuclear MMP-13 appears before other MMP activations

    • Suggests a potential initiating role in the MMP activation cascade

    • May represent an early therapeutic target window in stroke

This nuclear localization challenges the traditional view of MMPs as exclusively extracellular or membrane-associated enzymes and opens new avenues for understanding their roles in cellular responses to ischemia .

How do the multiple transcript variants of human MMP-13 differ in function and regulation?

The human MMP-13 gene generates several transcript variants, a unique feature among MMPs that adds complexity to its functional profile :

  • Known Transcript Variants:

    • Multiple mRNA species have been identified through Northern blot analysis

    • Variants differ in untranslated regions and potential coding sequences

    • May result from alternative promoter usage, splicing, or polyadenylation

  • Functional Implications:

    • Variants may encode proteins with altered:

      • Substrate specificity

      • Cellular localization (including nuclear targeting)

      • Activation mechanisms

      • Inhibitor sensitivity

    • Some variants might function as natural dominant-negative regulators

  • Differential Regulation:

    • Tissue-specific expression patterns of variants

    • Stimulus-specific induction (inflammatory vs. mechanical)

    • Developmental stage-specific regulation

    • Disease-associated shifts in variant profiles

  • Research Challenges:

    • Difficulty in specific detection of individual variants

    • Need for variant-specific antibodies or nucleic acid probes

    • Functional characterization requires expression of individual variants

    • Analysis of variant-specific interactions with substrates and inhibitors

Understanding these variant-specific differences could provide insights into the versatility of MMP-13 in different physiological and pathological contexts, potentially leading to more targeted therapeutic approaches .

What are the most promising approaches for selective MMP-13 inhibition in research and potential therapeutic applications?

Developing selective MMP-13 inhibitors presents significant challenges due to structural similarities with other MMPs, but several promising approaches have emerged:

  • Structure-Based Design Strategies:

    • Targeting the deep S1' pocket unique to MMP-13

    • Exploiting differences in zinc-binding groups and their orientation

    • Developing allosteric inhibitors that bind outside the catalytic site

    • Focusing on the hemopexin domain that influences substrate specificity

  • Biological Inhibitors:

    • Engineered variants of TIMPs with enhanced MMP-13 selectivity

    • Monoclonal antibodies against MMP-13-specific epitopes

    • Aptamers selected for MMP-13 binding

    • Modified substrates that act as competitive inhibitors

  • Transcriptional Regulation Approaches:

    • Targeting the AGRE negative regulatory element unique to MMP-13

    • Modulating specific transcription factors like those binding to OSE-2

    • Antisense oligonucleotides or siRNAs targeting MMP-13 mRNA

    • CRISPR-based transcriptional repression

  • Activation Interference:

    • Blocking the MMP-14/MMP-2 activation cascade specific to MMP-13

    • Peptides that mimic the propeptide domain to prevent activation

    • Compounds that stabilize the latent conformation

Inhibitor ApproachAdvantagesLimitationsCurrent Research Status
Catalytic Site InhibitorsDirect blocking of enzyme activityRisk of cross-reactivityMultiple candidates in preclinical testing
Allosteric InhibitorsHigher selectivity potentialMay have incomplete inhibitionEarly development phase
Antibody-BasedHigh specificityLimited tissue penetrationClinical trials for arthritis applications
Gene SilencingPrevents productionDelivery challengesProof-of-concept in animal models

These approaches offer pathways for developing research tools and potential therapeutics for conditions where MMP-13 plays a pathological role, such as arthritis, cancer, and cerebral ischemia .

What are the key considerations when designing experiments to study MMP-13 in human tissue samples?

When designing experiments to study MMP-13 in human tissues, researchers should consider several critical factors:

  • Sample Collection and Processing:

    • Rapid collection and appropriate preservation are essential (flash freezing for activity assays)

    • Include both pathological tissue and matched controls (e.g., contralateral brain regions in stroke)

    • Document clinical parameters and time from onset for acute conditions

    • Standardize processing protocols to maintain enzyme integrity

  • Detection Method Selection:

    • Activity vs. protein level measurement (often discordant)

    • Western blot can distinguish between latent (60kDa) and active (48kDa) forms

    • In situ zymography reveals spatial activity patterns

    • Immunohistochemistry identifies cellular sources and subcellular localization

  • Cellular Specificity Analysis:

    • Co-staining with cell-type markers (NeuN for neurons, GFAP for astrocytes)

    • Cell-type distribution varies between species (neurons in both humans and rats, oligodendrocytes in rats, astrocytes in humans)

    • Three-dimensional reconstruction to confirm intracellular localization

  • Specificity Controls:

    • Include broad-spectrum MMP inhibitors (e.g., 1,10-phenanthroline)

    • Antibody validation using MMP-13 knockout tissues or siRNA-treated samples

    • Distinguish from other collagenases (MMP-1, MMP-8) with similar substrate preferences

These methodological considerations ensure reliable and interpretable results when studying MMP-13 in human pathological specimens, particularly in conditions like stroke where timing and spatial distribution are critical factors .

How can researchers accurately distinguish between MMP-13 and other collagenases in experimental settings?

Accurate discrimination between MMP-13 and other collagenases (primarily MMP-1 and MMP-8) requires multiple complementary approaches:

  • Immunological Methods:

    • Use highly specific monoclonal antibodies validated against recombinant proteins

    • Confirm antibody specificity using western blots against purified MMP-1, MMP-8, and MMP-13

    • Employ epitope mapping to select antibodies targeting unique regions

    • Consider immunodepletion studies to confirm signal specificity

  • Substrate Preference Analysis:

    • MMP-13 has highest activity against type II collagen while MMP-1 preferentially cleaves type III collagen

    • Custom fluorogenic peptide substrates can exploit subtle differences in substrate preferences

    • Kinetic analysis (Km and kcat determinations) can quantify these differences

  • Inhibitor Profiling:

    • Differential sensitivity to TIMPs (TIMP-3 has higher affinity for MMP-13)

    • Test panels of selective chemical inhibitors with known MMP subtype profiles

    • Dose-response curves can reveal distinctive inhibition patterns

  • Molecular Biology Approaches:

    • RT-PCR with primers targeting unique sequences

    • Transcript analysis to detect MMP-13-specific splice variants

    • Knockdown or knockout validation to confirm specificity of signals

These approaches should be used in combination, as each method has limitations when used alone. This multi-modal strategy ensures reliable discrimination between these structurally similar yet functionally distinct collagenases in research applications .

What strategies can researchers employ to study the temporal dynamics of MMP-13 activation in disease progression?

Investigating the temporal dynamics of MMP-13 activation requires methodological approaches that capture activity changes across disease progression:

  • Time-Course Experimental Design:

    • In animal models: Sample collection at multiple timepoints (30min, 90min, 24h, 48h post-ischemia)

    • In patient cohorts: Longitudinal sampling when possible

    • Correlation with disease progression markers

    • Include pre-symptomatic stages when feasible

  • Live Imaging Approaches:

    • MMP-13-specific FRET (Förster Resonance Energy Transfer) probes

    • Activatable fluorescent probes that increase signal upon MMP-13 cleavage

    • Intravital microscopy in animal models

    • Correlative time-lapse imaging with functional outcomes

  • Activation State Assessment:

    • Antibodies specific to pro-domain or active forms

    • Ratio analysis of zymogen to active enzyme

    • Activity-based protein profiling with MMP-directed probes

    • Monitor activation intermediates in the MMP-14/MMP-2/MMP-13 cascade

  • Multi-Parameter Analysis:

    • Correlation with other MMPs in the activation cascade

    • Parallel assessment of inhibitors (TIMPs) and activators

    • Integration with broader proteomic and metabolomic data

    • Computational modeling of activation dynamics

The study by Cuadrado et al. demonstrates the importance of examining early timepoints, as MMP-13 activation was detected as early as 30 minutes after ischemia, preceding other MMPs and potentially initiating the activation cascade . This approach reveals MMP-13 as an early responder that may trigger subsequent pathological processes.

How might nuclear MMP-13 interact with chromatin and nuclear proteins in pathological conditions?

The discovery of nuclear MMP-13 localization opens intriguing questions about its potential interactions with nuclear components:

  • Potential Chromatin Interactions:

    • MMP-13 might process specific histones, affecting chromatin compaction

    • Could contribute to DNA fragmentation during apoptosis following ischemia

    • May modify chromatin-associated proteins that regulate gene access

    • Potential role in stress-induced chromatin reorganization

  • Nuclear Protein Substrates:

    • Poly-ADP-ribose polymerase (PARP) is a potential target during ischemia

    • Transcription factors might be processed to alter their activity

    • Nuclear matrix proteins could be modified, affecting nuclear architecture

    • DNA repair enzymes might be substrates, influencing cellular recovery potential

  • Transcriptional Regulation Involvement:

    • Direct or indirect effects on gene expression programs

    • Potential processing of transcriptional activators or repressors

    • Modification of nuclear receptors regulating stress responses

    • Creation of bioactive fragments with secondary signaling roles

  • Methodological Approaches to Study These Interactions:

    • Chromatin immunoprecipitation followed by mass spectrometry

    • Proximity labeling techniques in nuclear compartments

    • In vitro cleavage assays with nuclear protein fractions

    • Proteomic analysis of nuclear proteins following MMP-13 activation

This research direction could fundamentally alter our understanding of MMP biology, extending their roles from extracellular matrix remodeling to nuclear processes affecting gene expression and cell fate decisions during pathological conditions .

What role might extracellular vesicles play in the transport and activity of MMP-13 in the central nervous system?

Extracellular vesicles (EVs) represent an emerging area in MMP-13 biology with particular relevance to central nervous system pathologies:

  • MMP-13 Association with EVs:

    • MMP-13 may be packaged within different EV populations (exosomes, microvesicles)

    • Could be present as inactive pro-enzyme or active form

    • May be transported as cargo or associated with EV membranes

    • Selective enrichment in EVs from specific cell types (neurons, astrocytes, microglia)

  • Pathological Transport Mechanisms:

    • Neuronal EVs might deliver MMP-13 to distant brain regions

    • Glial-derived EVs could transfer MMP-13 to neurons during stress

    • EVs might cross the blood-brain barrier, facilitating peripheral-CNS communication

    • Could explain the differential cellular distribution observed in human vs. rat studies

  • Functional Consequences:

    • Protected transport of active MMP-13 avoiding inhibitor interactions

    • Targeted delivery to specific cell populations

    • Amplification of ischemic injury signals

    • Potential contribution to spreading pathology in neurodegenerative conditions

  • Experimental Approaches:

    • Isolation of EVs from brain tissue after ischemia

    • Characterization of MMP-13 content and activity in EV fractions

    • In vitro transfer studies using labeled EVs

    • Inhibition of EV formation to assess impact on MMP-13 distribution

This research direction could provide new insights into how MMP-13 activity spreads within the brain during pathological conditions like ischemia and offer novel therapeutic approaches targeting EV-mediated MMP-13 transport .

How can systems biology approaches advance our understanding of MMP-13 in complex disease networks?

Systems biology offers powerful frameworks for understanding MMP-13's role within complex disease networks:

  • Network Analysis Approaches:

    • Mapping MMP-13 interactions within the broader protease web

    • Identifying feedback loops in MMP activation cascades (MMP-14/MMP-2/MMP-13)

    • Quantifying the impact of MMP-13 perturbations on system-wide behavior

    • Integrating transcriptional regulatory networks controlling MMP-13 expression

  • Multi-Omics Integration:

    • Correlating MMP-13 activity with global proteome changes

    • Linking MMP-13 substrate processing to metabolomic alterations

    • Identifying compensatory mechanisms in MMP-13 deficiency

    • Discovering novel biomarkers associated with MMP-13 dysregulation

  • Computational Modeling:

    • Developing mathematical models of MMP-13 activation dynamics

    • Simulating effects of selective MMP-13 inhibition on disease progression

    • Predicting patient-specific responses based on molecular profiles

    • Optimizing combination therapy approaches targeting MMP-13 networks

  • Translational Applications:

    • Stratifying patients based on MMP-13 network signatures

    • Identifying optimal therapeutic intervention points within networks

    • Designing rational drug combinations affecting MMP-13 pathways

    • Developing personalized medicine approaches for conditions like stroke and arthritis

Systems approaches are particularly valuable given the complexity of MMP-13 regulation and its central position in MMP activation cascades . These methods can help resolve apparent contradictions in experimental data and provide more holistic understanding of MMP-13's role across different pathologies.

What are the most significant recent advances in our understanding of human MMP-13 biology?

The most significant recent advances in human MMP-13 biology include:

  • Nuclear Localization and Function:

    • Discovery of MMP-13 nuclear translocation after ischemic stimulus

    • Identification of putative nuclear localization signals in MMP-13 sequence

    • Evidence of nuclear gelatinolytic activity in neural cells

    • Implications for direct roles in nuclear processes during cell stress

  • Regulatory Complexity:

    • Identification of multiple transcript variants unique to MMP-13

    • Discovery of the negative regulatory element AGRE in the MMP-13 promoter

    • Characterization of the MMP-14/MMP-2/MMP-13 activation cascade

    • Elucidation of tissue-specific and pathology-specific regulatory mechanisms

  • Cellular Distribution Patterns:

    • Species-specific differences in cellular sources (neurons in both humans and rats, oligodendrocytes in rats, astrocytes in humans)

    • Temporal dynamics of expression in pathological conditions

    • Restricted distribution in normal tissues versus selective upregulation in disease

  • Early Role in Pathological Cascades:

    • Evidence for MMP-13 as one of the earliest MMPs activated after ischemia (30 minutes)

    • Potential initiating role in broader MMP activation sequences

    • Implications for therapeutic targeting windows in acute conditions

These advances collectively reveal MMP-13 as a more complex and versatile enzyme than originally thought, with functions potentially extending beyond extracellular matrix remodeling to include nuclear processes and early signaling in pathological cascades .

What are the major unresolved questions that should drive future research on human MMP-13?

Despite significant advances, several critical questions about human MMP-13 remain unresolved:

  • Nuclear Function Mechanisms:

    • What are the specific nuclear substrates of MMP-13?

    • How does nuclear MMP-13 affect gene expression and chromatin structure?

    • What triggers nuclear translocation and how is it regulated?

    • Does nuclear MMP-13 contribute to cell fate decisions during stress?

  • Transcript Variant Functions:

    • What are the functional differences between MMP-13 transcript variants?

    • How are these variants differentially regulated in health and disease?

    • Do variants have distinct subcellular localizations or substrate preferences?

    • Can variant-specific targeting offer therapeutic advantages?

  • Therapeutic Targeting Challenges:

    • How can truly selective MMP-13 inhibition be achieved?

    • What are the optimal therapeutic windows for MMP-13 inhibition in acute conditions?

    • Are there disease-specific MMP-13 functions that could be selectively targeted?

    • What combination approaches might overcome limitations of single-target inhibition?

  • Physiological Roles:

    • What are the normal physiological functions of MMP-13 in the brain and other tissues?

    • How does MMP-13 contribute to normal tissue homeostasis and repair?

    • What compensatory mechanisms exist when MMP-13 is absent or inhibited?

    • How does aging affect MMP-13 expression and function?

Addressing these questions will require interdisciplinary approaches combining structural biology, systems biology, advanced imaging, and translational research to fully understand MMP-13's complex roles in human health and disease .

How might emerging technologies advance our ability to study and target MMP-13 in research and clinical applications?

Emerging technologies offer exciting opportunities to advance MMP-13 research and clinical applications:

  • Single-Cell Technologies:

    • Single-cell RNA sequencing to identify cell populations expressing MMP-13 variants

    • Single-cell proteomics to detect cell-specific MMP-13 activation patterns

    • Spatial transcriptomics to map MMP-13 expression in tissue microenvironments

    • These approaches could resolve the heterogeneity in cellular sources between species

  • Advanced Imaging Techniques:

    • Super-resolution microscopy for precise subcellular localization

    • Intravital imaging with MMP-13-specific activity probes

    • Correlative light and electron microscopy to connect function with ultrastructure

    • PET imaging with MMP-13-selective tracers for clinical translation

  • CRISPR-Based Technologies:

    • Precise genome editing to study specific domains and variants

    • CRISPRi/CRISPRa for temporal control of MMP-13 expression

    • Base editing to introduce or correct disease-associated variants

    • In vivo CRISPR screens to identify MMP-13 regulators

  • Therapeutic Development Platforms:

    • DNA-encoded libraries for discovering selective inhibitors

    • Antibody engineering for enhanced tissue penetration

    • Nanobody development for accessing restricted domains

    • Targeted protein degradation approaches (PROTACs) for selective MMP-13 removal

  • Artificial Intelligence Applications:

    • Machine learning for predicting MMP-13 substrates and interactions

    • Deep learning analysis of complex multi-omics datasets

    • AI-assisted drug design for selective inhibitors

    • Predictive modeling of patient responses to MMP-13-targeting therapies

Product Science Overview

Introduction

Matrix Metalloproteinase-13 (MMP-13), also known as collagenase-3, is a member of the matrix metalloproteinase (MMP) family. These enzymes are involved in the breakdown of extracellular matrix (ECM) components, which is crucial for various physiological processes such as tissue remodeling, wound healing, and embryonic development. MMP-13 is particularly known for its ability to degrade collagen, a major structural protein in the ECM.

Structure and Expression

MMP-13 is a zinc-dependent endopeptidase that is synthesized as a proenzyme (proMMP-13) and requires activation to become fully functional. The human recombinant form of MMP-13 is typically expressed in Escherichia coli (E. coli) and purified using chromatographic techniques. The recombinant protein often includes a His-tag to facilitate purification and detection .

Function and Mechanism

MMP-13 plays a significant role in the degradation of various types of collagen, including type I, II, III, IV, X, and XIV. It has the highest activity with soluble type II collagen. The enzyme is involved in several physiological and pathological processes:

  • Tissue Remodeling: MMP-13 is essential for the remodeling of the ECM during development and wound healing. It helps in the turnover of collagen and other matrix components.
  • Bone Development: MMP-13 is required for normal embryonic bone development and ossification. It plays a role in the healing of bone fractures via endochondral ossification.
  • Cartilage Degradation: MMP-13 is implicated in the degradation of cartilage in diseases such as osteoarthritis. It promotes the irreversible destruction of cartilage collagen .
Clinical Significance

Due to its role in ECM degradation, MMP-13 is a target for therapeutic intervention in various diseases. Inhibitors of MMP-13 are being explored for the treatment of conditions like osteoarthritis, where excessive collagen degradation leads to joint damage. Additionally, MMP-13 is being studied for its role in cancer metastasis, as it can facilitate tumor cell invasion by degrading the ECM .

Recombinant MMP-13

Recombinant human MMP-13 is used extensively in research to study its structure, function, and role in various diseases. The recombinant protein is typically produced in E. coli and purified to high purity levels (>95%) for use in biochemical assays and structural studies. It is available in both proenzyme and active forms, allowing researchers to investigate the activation mechanisms and enzymatic activity of MMP-13 .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.