S100A4 (S100 Calcium-Binding Protein A4) is a member of the S100 family, characterized by two EF-hand calcium-binding motifs. It exists as a symmetric homodimer stabilized by noncovalent interactions and is encoded by the S100A4 gene located on chromosome 1q21 . While physiologically involved in neurogenesis and cell differentiation, its dysregulation is linked to cancer metastasis, chronic inflammation, and tissue fibrosis .
S100A4 is a key driver of tumor invasiveness and metastasis through multiple mechanisms:
Metastasis Promotion: Overexpression enhances tumor cell migration by regulating cytoskeletal dynamics (e.g., binding myosin IIA) and inducing matrix metalloproteinases (MMPs) .
Angiogenesis: Synergizes with VEGF to increase endothelial cell migration and MMP-9 activity .
Biomarker Potential: Elevated expression correlates with poor prognosis in prostate, breast, and pancreatic cancers .
S100A4 contributes to chronic inflammatory and fibrotic diseases:
Periapical Granulomas: Upregulates IL-1β and MMPs, exacerbating bone destruction in oral inflammatory lesions .
Fibrosis: Drives collagen deposition in liver, lung, and synovial tissues .
Autoimmune Diseases: Promotes macrophage chemotaxis and cytokine secretion in rheumatoid arthritis .
S100A4 binds myosin IIA, inhibiting phosphorylation of myosin heavy chains (MHC) and disrupting filament assembly, which enhances cell motility .
MMP-9 Activation: S100A4 transcriptionally upregulates MMP-9 and TIMP-1, facilitating extracellular matrix degradation .
In Vivo Evidence: Silencing S100A4 in prostate cancer xenografts reduced tumor growth by >50% over 21 weeks .
Secreted S100A4 recruits immune cells (e.g., macrophages, T-lymphocytes) and stimulates pro-inflammatory cytokines like TNF-α .
Antibody Therapy: Monoclonal antibody 5C3 neutralizes extracellular S100A4, reducing tumor growth by 70% in melanoma models .
Gene Silencing: siRNA-mediated knockdown inhibits MMP-9 activity and metastasis in prostate cancer .
S100A4 is a member of the S100 family of calcium-binding proteins located on chromosome 1q21 in humans. The protein is 101 amino acids in length with a molecular mass of approximately 11.5 kDa. Its structure is characterized by two calcium-binding EF-hand domains, with the first EF-hand located between amino acids 12-47 . Upon calcium binding, S100A4 undergoes a conformational change that forms a hydrophobic pocket essential for target protein recognition . The human S100A4 gene consists of four exons, with the first two being non-coding . This structural arrangement enables S100A4 to participate in both calcium-dependent and calcium-independent protein-protein interactions that are crucial for its various biological functions.
Contrary to the previously held belief that S100A4 expression is restricted to fibroblasts, research has demonstrated that S100A4 is expressed in a diverse range of normal cell types. These include:
Immune cells: monocytes, macrophages, and T lymphocytes
Neuronal subpopulations in the brain
Certain populations in the developing nervous system
Various cells in the hippocampus and temporal cortex with a specific spatio-temporal pattern of expression
S100A4 serves multiple physiological functions in normal tissues:
Neural development: S100A4 contributes to nervous system development with a specific spatio-temporal expression pattern in the human hippocampus and temporal cortex that declines with aging .
Myelination: S100A4 immunoreactivity appears in newly myelinated areas postnatally, suggesting a role in the maturation of myelinated fiber tracts .
Neuronal survival and plasticity: Extracellular S100A4, particularly in oligomeric quaternary structures, promotes neurite outgrowth and survival after apoptotic stimuli in hippocampal, dopaminergic, and cerebellar neurons .
Cell motility and differentiation: S100A4 regulates cellular motility, chemotaxis, and differentiation processes essential for normal tissue homeostasis .
Cell-to-cell communication: The protein facilitates intercellular signaling, particularly in the nervous system .
Aging processes: S100A4 has been found in corpora amylacea (hyaline structures associated with normal aging), suggesting a function in senescence-associated inflammation .
Understanding these normal functions provides context for how S100A4 dysregulation contributes to pathological conditions.
S100A4 facilitates cancer metastasis through multiple interconnected molecular mechanisms:
MMP regulation: S100A4 controls the invasive potential of cancer cells by regulating matrix metalloproteinase 9 (MMP-9) expression and activity. Suppression of S100A4 by siRNA significantly reduces MMP-9 expression, proteolytic activity, and transcriptional activation, while S100A4 overexpression produces the opposite effect .
TIMP-1 modulation: S100A4 also influences the expression of tissue inhibitor of metalloproteinase 1 (TIMP-1), which responds to S100A4 gene suppression, suggesting a complex regulatory relationship between S100A4 and the MMP/TIMP balance .
Cellular proliferation control: S100A4 influences the proliferative capacity of cancer cells. siRNA-mediated suppression of S100A4 significantly reduces the proliferative potential of prostate cancer cells .
Transcriptional regulation: S100A4 affects the transcriptional activation of genes involved in invasion and metastasis, as demonstrated by reduced transcriptional activation of MMP-9 in S100A4-siRNA-transfected cells .
In vivo tumor growth promotion: S100A4 enhances tumor growth in vivo. Mice implanted with S100A4-siRNA-transfected prostate cancer cells showed significantly reduced tumor growth rates and increased tumor-free survival compared to controls .
These molecular mechanisms collectively contribute to S100A4's potent metastasis-promoting effects, making it a significant biomarker and potential therapeutic target in cancer.
S100A4 functions as a Damage-Associated Molecular Pattern (DAMP) protein that plays a critical role in fibrotic diseases, particularly systemic sclerosis (SSc):
Overexpression in fibrotic conditions: S100A4 is overexpressed in patients with SSc, with levels correlating with organ involvement and disease activity .
Dual inflammatory and fibrotic roles: Research demonstrates that S100A4 promotes both pro-inflammatory phenotypes and organ pro-fibrotic pathways in multiple tissues including the liver, kidney, lung, heart, tendons, and synovial tissues .
Protective effect of S100A4 deficiency: S100A4-deficient mice (S100A4^-/-) show protection from fibrosis development, indicating a causal role in fibrotic processes .
Anti-S100A4 monoclonal antibodies (mAbs) have emerged as a promising therapeutic strategy:
In a bleomycin-induced skin fibrosis model, anti-S100A4 mAbs effectively reduced dermal thickening, myofibroblast counts, and collagen accumulation .
Transcriptional profiling revealed that S100A4 inhibition targets multiple profibrotic and proinflammatory processes relevant to SSc pathogenesis .
Ex vivo studies using precision-cut SSc skin slices demonstrated that S100A4 inhibition modulates inflammation- and fibrosis-relevant gene sets .
Downstream targets of S100A4, including AMP-activated protein kinase, calsequestrin-1, and phosphorylated STAT3, have been validated, with STAT3 inhibition preventing S100A4's profibrotic effects on fibroblasts in human skin .
These findings support the development of anti-S100A4 mAbs as disease-modifying targeted therapies for fibrotic diseases, offering dual targeting of inflammatory and fibrotic pathways.
S100A4 exhibits distinct signaling mechanisms depending on its localization:
Calcium-dependent conformational changes enable interactions with cytoskeletal components
Regulates gene expression through direct or indirect interactions with transcription factors
Affects MMP-9 gene transcriptional activation as demonstrated in transfection studies
Interacts with cytoskeletal proteins affecting cellular motility and structural organization
Functions as a secreted signaling molecule that can act on various cell types expressing appropriate receptors
Promotes neurite outgrowth and neuronal survival in a RAGE-independent fashion
Most effective when arranged in oligomeric quaternary structures that interact with heparan sulfate proteoglycans on neuronal cell surfaces
Released into biological fluids in various cancer types, suggesting potential as a biomarker for early-stage tumors and metastatic events
Activates intracellular signaling cascades, including calcium mobilization pathways, in target cells
This dual functionality allows S100A4 to mediate both intracellular processes and cell-to-cell communication, expanding its influence beyond the cells that produce it to affect neighboring cells and tissues in both physiological and pathological contexts.
Research on S100A4's role in cancer metastasis can be effectively approached through these complementary methods:
siRNA-mediated gene suppression: Transfection of cancer cell lines (e.g., PC-3 prostate cancer cells) with S100A4-specific siRNA effectively reduces S100A4 expression, allowing assessment of invasion and proliferation capabilities .
Overexpression studies: Transfection with pcDNA3.1-S100A4 plasmid enables examination of gain-of-function effects on metastatic potential .
Invasion assays: Matrigel-based invasion chambers to quantify the invasive capability of cells with altered S100A4 expression .
Proliferation assays: BrdU incorporation or MTT assays to measure cell proliferation rates .
MMP activity assays: Gelatin zymography to assess MMP-9 proteolytic activity in relation to S100A4 expression levels .
Macroarray screening: Using arrays containing metastasis-related genes (e.g., 96 well-characterized metastatic genes) to identify downstream targets of S100A4 .
Promoter-reporter assays: Employing MMP-9-promoter reporters to evaluate transcriptional activation in response to S100A4 modulation .
Subcutaneous xenograft models: Implantation of S100A4-manipulated cancer cells into nude mice to evaluate tumor growth rates and tumor-free survival .
Kaplan-Meier survival analysis: Assessment of tumor-free survival to determine the impact of S100A4 on tumor development and progression .
Immunohistochemical staining of patient samples to correlate S100A4 expression with clinical outcomes and metastatic status .
Prognostic value assessment through long-term patient follow-up studies (e.g., 19-year follow-up cohorts) .
This multi-modal approach provides comprehensive insights into both molecular mechanisms and clinical relevance of S100A4 in cancer metastasis.
Optimal techniques for evaluating S100A4 expression in clinical samples include:
Gold standard for clinical samples due to preservation of tissue architecture
Requires careful antibody validation using multiple antibodies (both monoclonal and polyclonal) to avoid cross-reactivity with other S100 family members
Critical consideration of fixation methods, as different methods can impact results and lead to conflicting findings in studies
Enables correlation with clinical outcomes when combined with long-term patient follow-up data
RNA sequencing of clinical samples or precision-cut tissue slices to evaluate S100A4 mRNA expression
RT-PCR to quantify S100A4 mRNA levels, particularly useful for confirming protein expression data in specific cell types
In situ hybridization to localize S100A4 mRNA expression within the tissue context
Western blotting for quantitative assessment of S100A4 protein levels
Proteomic approaches such as shotgun proteomics, which have been used to identify S100A4 in meningioma biopsies with differential expression between male and female patients
ELISA or other immunoassays to measure S100A4 levels in biological fluids (serum, plasma, or other disease-relevant fluids)
Important for evaluating S100A4 as a non-invasive biomarker for early tumor detection or monitoring metastatic events
Include positive and negative controls when assessing S100A4 expression
Validate results using multiple techniques when possible
Consider cell-type specific expression patterns rather than assuming fibroblast-restricted expression
These techniques provide complementary information about S100A4 expression, with the selection depending on the specific research question, sample availability, and required level of molecular detail.
To effectively study S100A4's dual role in inflammation and fibrosis, researchers should design experiments that address both pathways simultaneously:
Animal models: Use established fibrosis models (e.g., bleomycin-induced skin fibrosis and Tsk-1 mice) with therapeutic dosing regimens of anti-S100A4 antibodies .
Ex vivo models: Employ precision-cut tissue slices from affected organs of patients (as used with SSc skin) to preserve the complex cellular interactions in the native tissue microenvironment .
Comparative studies: Include both inflammatory models (e.g., LPS challenge) and fibrotic models to distinguish pathway-specific effects.
Prevention vs. regression studies: Design experiments with different intervention timelines to assess both prevention of fibrosis development and regression of pre-established fibrosis .
Therapeutic dosing regimens: Administer treatments after disease establishment to mirror clinical scenarios .
Transcriptional profiling: Perform RNA sequencing to identify both inflammation- and fibrosis-relevant gene sets affected by S100A4 modulation .
Protein validation: Confirm transcriptional changes at the protein level for key downstream targets (e.g., AMP-activated protein kinase, calsequestrin-1, STAT3) .
Histological assessment: Quantify dermal thickening, myofibroblast counts, and collagen accumulation to evaluate fibrotic changes .
Inflammatory markers: Measure cytokine/chemokine profiles and immune cell infiltration to assess inflammatory components.
Pathway inhibition studies: Use specific inhibitors (e.g., STAT3 inhibitors) to block downstream effectors and determine their contribution to S100A4's effects .
Cell-specific knockouts: Generate conditional S100A4 knockout models in specific cell populations (fibroblasts vs. immune cells) to delineate cell-specific contributions.
Receptor blockade experiments: Block specific S100A4 receptors to determine which signaling pathways mediate inflammatory versus fibrotic responses.
Human sample correlation: Correlate experimental findings with S100A4 expression patterns in patient samples, disease activity, and organ involvement .
Biomarker development: Evaluate whether S100A4 or its downstream effectors can serve as biomarkers for both inflammatory and fibrotic disease components.
This comprehensive experimental approach enables researchers to untangle the complex dual role of S100A4 in inflammation and fibrosis, facilitating the development of targeted therapeutic strategies.
When faced with contradictory findings regarding S100A4 expression and patient outcomes, researchers should adopt this systematic analysis approach:
Fixation methods: Different tissue fixation protocols can significantly affect S100A4 detection. For example, conflicting results in breast cancer studies have been attributed to variations in fixation methods .
Antibody selection: S100 proteins share homology, and antibody cross-reactivity may lead to inconsistent results. Validate findings using multiple antibodies (both monoclonal and polyclonal) .
Detection techniques: Compare IHC, Western blotting, and mRNA-based methods, as each has different sensitivity and specificity profiles.
Patient population heterogeneity: Analyze how differences in age, gender, ethnicity, and comorbidities might influence results.
Cancer stage distribution: Variations in the proportion of early versus late-stage patients can dramatically affect outcome correlations. In some studies, conflicting results were attributed to differences in stage distribution .
Follow-up duration: Longer observation periods (e.g., 19 years in Rudland's breast cancer study) may reveal associations not apparent in shorter studies .
Cancer type specificity: S100A4's prognostic value may vary between cancer types due to tissue-specific molecular contexts.
Cell type expression patterns: Consider which cells are expressing S100A4 (tumor cells vs. stromal cells) and how this might affect interpretation.
S100A4 subcellular localization: Distinguish between nuclear, cytoplasmic, and extracellular S100A4, as each may have different functional implications.
Sample size adequacy: Smaller studies may lack statistical power to detect associations. Some contradictory S100A4 studies had notably different sample sizes .
Statistical methods: Different statistical approaches to survival analysis may yield varying results.
Multivariate vs. univariate analysis: Determine whether confounding variables were adequately controlled for.
Threshold effects: Consider whether S100A4 might have non-linear effects on outcomes that manifest only above certain expression thresholds.
Interaction effects: Investigate whether S100A4's prognostic value depends on its interaction with other molecular markers.
Meta-analysis approach: When possible, perform meta-analyses to integrate results across studies and identify factors explaining heterogeneity.
By systematically addressing these dimensions, researchers can better interpret apparently contradictory findings and develop a more nuanced understanding of S100A4's role in cancer progression and patient outcomes.
When analyzing S100A4's role across different tissues and pathologies, researchers should consider:
Baseline expression patterns: S100A4 shows tissue-specific and developmental stage-specific expression patterns. In the nervous system, it displays a spatio-temporal pattern in the hippocampus and temporal cortex that changes with development and aging .
Cell type specificity: Contrary to earlier beliefs, S100A4 is expressed in various cell types beyond fibroblasts, including immune cells and neuronal subpopulations . This diverse expression must be considered when interpreting tissue-specific functions.
Physiological roles: S100A4's normal functions vary by tissue, from neurogenesis and myelination in neural tissues to roles in cellular motility and differentiation in other tissues.
Cancer vs. fibrosis vs. neurodegeneration: S100A4 may operate through distinct pathways in different conditions. In cancer, it regulates MMP-9 to promote invasion , while in fibrosis, it activates STAT3 and other fibrotic pathways .
Intracellular vs. extracellular functions: S100A4 can function both intracellularly and as a secreted factor, with different mechanisms predominating in different pathologies .
Interacting partners: Identify tissue-specific binding partners and signaling pathways activated by S100A4 in each context.
Cross-tissue models: Develop experimental designs that compare S100A4 functions across multiple tissue types under controlled conditions.
Multiple disease models: Study S100A4 in parallel models of cancer, fibrosis, and neurological conditions to identify common and distinct mechanisms.
Temporal dynamics: Evaluate how S100A4's role changes during disease progression in different tissues.
Biomarker potential across conditions: Assess whether S100A4 has similar prognostic/diagnostic value across different pathologies. For example, it strongly predicts outcomes in breast cancer but may have different implications in neurodegenerative diseases .
Therapeutic targeting considerations: Determine whether inhibition strategies effective in one disease context (e.g., anti-S100A4 mAbs in fibrosis ) would be applicable in others.
Potential side effects: Consider how targeting S100A4 in one disease might affect its physiological functions in other tissues.
Detection protocols: Develop standardized protocols for S100A4 detection that can be applied across different tissues and conditions.
Quantification approaches: Establish consistent methods for quantifying S100A4 expression and activity that allow for cross-tissue and cross-disease comparisons.
Data integration strategies: Implement computational approaches to integrate S100A4 data across different tissues and pathologies to identify global patterns.
By systematically addressing these considerations, researchers can develop a comprehensive understanding of S100A4's diverse roles and identify both common mechanisms and tissue-specific functions relevant to targeted therapeutic development.
Distinguishing correlation from causation in S100A4 research requires methodological rigor across several dimensions:
Knockdown/knockout studies: The observation that S100A4^-/- mice are protected from fibrosis development provides strong evidence for causality in fibrotic processes . Similarly, siRNA-mediated suppression of S100A4 significantly reducing cancer cell invasiveness supports a causal role in metastasis .
Overexpression models: Cells transfected with pcDNA3.1-S100A4 plasmid demonstrating increased MMP-9 expression and enhanced invasiveness provide complementary evidence for causality .
Rescue experiments: Reintroducing S100A4 into knockout systems and observing restoration of phenotypes provides convincing evidence of direct causality.
Time-course analyses: Studying S100A4 expression changes preceding phenotypic changes helps establish directional relationships.
Inducible systems: Using temporally controlled gene expression/suppression systems to manipulate S100A4 at different disease stages.
Longitudinal studies: In human research, conducting prospective studies rather than cross-sectional analyses strengthens causal inferences.
Titration experiments: Demonstrating proportional relationships between S100A4 levels and outcome measures supports causality.
Partial knockdown studies: Showing that varying degrees of S100A4 suppression lead to proportional changes in phenotypes.
Concentration-dependent effects: Assessing how different concentrations of recombinant S100A4 affect cellular responses in vitro.
Downstream target identification: The identification of specific targets like MMP-9 that respond to S100A4 manipulation helps establish mechanistic causality .
Pathway inhibition: Demonstrating that STAT3 inhibition prevents S100A4's profibrotic effects confirms this pathway as a causal mediator .
Molecular interaction studies: Biochemical validation of direct interactions between S100A4 and proposed effector molecules.
Target-specific interventions: The effectiveness of anti-S100A4 monoclonal antibodies in reducing established fibrosis strongly supports a causal role .
Therapeutic response correlation: Showing that clinical improvement correlates with changes in S100A4-regulated pathways after treatment.
Combination approaches: Demonstrating synergistic effects between S100A4 inhibition and other pathway-specific interventions.
Consistent findings across species: When both human correlative studies and animal causal studies show aligned results, causality is more strongly supported.
Ex vivo validation: Using precision-cut human tissue slices to validate mechanisms observed in animal models strengthens causal claims .
Genetic association studies: Identifying S100A4 genetic variants associated with disease risk or progression provides additional support for causal relationships.
By integrating these methodological approaches and critically evaluating the strength of evidence across multiple experimental systems, researchers can build a compelling case for causal relationships between S100A4 and disease processes, moving beyond mere correlation.
Several cutting-edge technologies and approaches are poised to transform S100A4 research:
Super-resolution microscopy: To visualize S100A4's subcellular localization and dynamic interactions with binding partners at nanoscale resolution.
Intravital imaging: For real-time visualization of S100A4-expressing cells in living tissues during disease progression and treatment response.
Multiparametric imaging: Combining S100A4 detection with multiple markers to understand its relationship with the tissue microenvironment.
Single-cell RNA sequencing: To identify cell-specific expression patterns of S100A4 and its downstream targets across heterogeneous tissues.
Single-cell proteomics: For comprehensive protein-level analysis of S100A4 signaling networks in individual cells.
Spatial transcriptomics: To map S100A4 expression patterns within tissue architectural contexts, revealing spatial relationships with other cell types.
CRISPR screening: Genome-wide or targeted screens to identify novel regulators and effectors of S100A4 signaling.
CRISPR base editing: For precise modification of S100A4 regulatory elements to understand transcriptional control mechanisms.
CRISPR-mediated lineage tracing: To track the fate of S100A4-expressing cells during disease progression and treatment.
Cryo-electron microscopy: To determine high-resolution structures of S100A4 in complex with its binding partners and potential therapeutic agents.
Proximity labeling proteomics: BioID or APEX approaches to comprehensively map S100A4's protein interaction networks in living cells.
Hydrogen-deuterium exchange mass spectrometry: To characterize dynamic conformational changes in S100A4 upon calcium binding and target recognition.
Organoid technologies: Patient-derived 3D organoids to study S100A4 functions in physiologically relevant systems.
Microphysiological systems (organ-on-chip): To model complex tissue interactions and S100A4-mediated processes under controlled conditions.
Antibody engineering: Development of next-generation anti-S100A4 antibodies with enhanced tissue penetration and effector functions.
Network medicine approaches: To position S100A4 within broader disease networks and identify potential combination therapy targets.
Machine learning algorithms: For integration of multi-omics data to predict S100A4-dependent disease trajectories and treatment responses.
Molecular dynamics simulations: To understand how S100A4 conformational changes mediate its diverse functions and identify potential druggable pockets.
These emerging technologies will enable researchers to address fundamental questions about S100A4 biology with unprecedented resolution and scale, potentially leading to breakthrough discoveries and novel therapeutic strategies.
The most promising therapeutic avenues for targeting S100A4 include:
Disease-modifying antibodies: Anti-S100A4 mAbs have shown efficacy in treating pre-established fibrosis and in regression of existing fibrosis in animal models, demonstrating their potential as disease-modifying agents .
Dual-targeting potential: These antibodies uniquely target both inflammatory and fibrotic pathways, making them particularly valuable for complex diseases like systemic sclerosis .
Format optimization: Development of various antibody formats (full IgG, Fab fragments, bispecifics) optimized for specific tissue penetration and pharmacokinetics.
Structure-based design: Using the known structure of S100A4's calcium-binding EF-hands and hydrophobic pocket to design small molecules that interfere with target protein recognition .
Allosteric modulators: Development of compounds that alter S100A4's conformational changes in response to calcium, thereby modulating its function.
Pathway-specific inhibitors: Targeting downstream effectors like STAT3, which has been validated as mediating S100A4's profibrotic effects in human skin .
siRNA delivery systems: Building on the successful S100A4 silencing achieved in experimental models , developing targeted delivery systems for siRNA to specific tissues.
Antisense oligonucleotides: Design of antisense therapies to reduce S100A4 expression in affected tissues.
mRNA vaccination: Exploration of mRNA-based approaches to generate immune responses against cells overexpressing S100A4.
S100A4 and MMP inhibition: Combining S100A4 targeting with MMP-9 inhibitors to enhance anti-metastatic effects, given the established regulatory relationship between these molecules .
Multi-pathway targeting: Developing combination therapies that simultaneously target S100A4 and complementary pathways relevant to specific diseases.
Sequential therapy approaches: Strategically timing S100A4 inhibition with other treatments to maximize therapeutic efficacy.
Companion diagnostics: Developing S100A4 expression assays to identify patients most likely to benefit from S100A4-targeted therapies.
Response prediction: Using S100A4 levels in biological fluids as a biomarker to monitor treatment response .
Personalized dosing strategies: Tailoring treatment intensity based on individual patient S100A4 expression patterns.
Cancer metastasis prevention: Using S100A4 inhibitors as adjuvant therapy in high-risk cancer patients to prevent metastatic spread .
Fibrosis regression: Applying anti-S100A4 therapies to reverse established fibrosis in systemic sclerosis and other fibrotic conditions .
Neurological applications: Exploring the potential of S100A4-targeting strategies in neurodegenerative conditions where S100A4 dysregulation has been implicated .
The convergence of these therapeutic approaches with advanced understanding of S100A4 biology presents transformative opportunities for treating multiple diseases where S100A4 plays a pathogenic role.
S100 Calcium-Binding Protein A4 (S100A4), also known as metastasin, is a member of the S100 family of proteins. These proteins are characterized by their ability to bind calcium ions through EF-hand motifs, which are helix-loop-helix structural domains . S100A4 is encoded by the S100A4 gene located on chromosome 1q21 in humans .
S100A4 is a small protein consisting of 101 amino acids . It contains two EF-hand calcium-binding motifs, which are crucial for its function. The protein is localized in the cytoplasm and/or nucleus of various cell types and is involved in several cellular processes, including cell motility, invasion, and tubulin polymerization .
S100A4 plays a significant role in the regulation of the cell cycle, differentiation, and apoptosis. It is also involved in the epithelial to mesenchymal transition (EMT), a process critical for cancer metastasis . The protein interacts with non-muscle myosin heavy chain IIA (NMMHC IIA), enhancing cell motility and invasiveness .
The expression of S100A4 is often upregulated in various cancers, including breast, colorectal, and pancreatic cancers . Its overexpression is associated with poor prognosis and increased metastatic potential. S100A4 has been shown to interact with other proteins, such as S100A1, and is implicated in tumor progression and metastasis .
Given its role in cancer progression, S100A4 is considered a potential therapeutic target. Inhibiting its expression or function could potentially reduce tumor invasiveness and metastasis . Research is ongoing to develop specific inhibitors that can target S100A4 and its interactions with other proteins.