Regulation by Tumor Suppressors/Oncogenes:
Transcription Factors:
Polymorphisms:
Anti-angiogenic: Inhibits endothelial cell migration via CD36 binding and TGFβ1 activation .
Pro-angiogenic in Follicles: Promotes ovarian follicle capillary sprouting by enhancing endothelial tip cell migration .
Suppresses T-cell activation by reducing CD69, IFNγ, and TCF7 expression .
Increases CTLA4+CD8+ T cells, fostering an immunosuppressive tumor microenvironment (TME) .
THBS1 exhibits dual roles depending on context:
Paradoxical Effects:
Thrombospondin-1 (TSP1) is a homotrimeric glycoprotein and a member of the Thrombospondin family. It is encoded by the THBS1 gene. TSP1 is involved in cell-to-cell and cell-to-matrix interactions. It binds to various molecules, including fibrinogen, fibronectin, laminin, type V collagen, and integrins alpha-V/beta-1. TSP1 plays a crucial role in platelet aggregation, angiogenesis, and tumorigenesis. It acts as an inhibitor of neovascularization and tumorigenesis in healthy tissues. TSP1 interacts with numerous cell adhesion receptors, such as CD36, av integrins, b1 integrins, syndecan, and integrin-associated protein (IAP or CD47). Additionally, it interacts with proteases involved in angiogenesis, including plasminogen, matrix metalloproteinase, thrombin, cathepsin, and elastase. TSP1 exhibits both positive and negative modulatory effects on endothelial cell adhesion, motility, and growth. Recent studies have revealed that TSP1 binds to reelin receptors, ApoER2 and VLDLR, influencing neuronal migration in the rostral migratory stream.
THBS1 is a matricellular protein that regulates cell migration through direct binding interactions with integrin proteins and other receptors including CD47 and CD36. It plays crucial roles in tissue remodeling and wound repair, particularly through activation of transforming growth factor-β1 (TGF-β1) . Research methodologies to study basic THBS1 function typically involve gene expression analysis through techniques like qPCR, in situ hybridization (RNAscope), and immunohistochemistry to visualize THBS1 distribution in tissues . THBS1 has seemingly contradictory functions as both an endogenous inhibitor of angiogenesis and a promoter of tumor invasion and metastasis in certain contexts .
THBS1 transcription is regulated through multiple pathways, with TGF-β signaling being particularly important. Research has demonstrated that the TGF-β canonical pathway transcriptionally regulates THBS1 expression through SMAD3 binding to the THBS1 gene promoter . This creates a positive feedback loop, as THBS1 can also activate TGF-β1 . To study this regulation experimentally, researchers often employ promoter analysis, chromatin immunoprecipitation (ChIP) assays to confirm SMAD3 binding, and reporter gene assays to measure promoter activity under different conditions.
THBS1 demonstrates complex and sometimes contradictory roles in cancer biology. Global expression analyses have revealed that THBS1 is upregulated in high-grade gliomas and associated with poor prognosis . Similarly, in gastric cancer, THBS1 is overexpressed and correlates with poor patient outcomes, with high THBS1 expression identified as an independent risk factor for survival . Methodologically, researchers assess THBS1's contribution to cancer through a combination of clinical correlation studies, gene set enrichment analysis (GSEA), and functional assays examining cell migration, invasion, and angiogenesis in response to THBS1 manipulation.
THBS1 expression significantly influences the tumor immune microenvironment. Research has demonstrated that THBS1 expression is most strongly correlated with tumor-associated macrophages (TAMs), M2 macrophages, and cancer-associated fibroblasts (CAFs) in gastric cancer . Moreover, THBS1 expression positively correlates with most immune checkpoint members, suggesting it plays an important role in immunosuppression within the tumor microenvironment . To investigate these relationships, researchers use techniques such as the ESTIMATE algorithm for immune infiltration analysis, single-cell transcriptome analysis, and CIBERSORT algorithm to deconvolute immune cell populations .
THBS1 overexpression has been negatively correlated with sensitivity to certain anticancer drugs. For example, in gastric cancer, elevated THBS1 levels were associated with decreased sensitivity to oxaliplatin . This suggests that THBS1 might be a marker for chemoresistance and potentially a therapeutic target to enhance treatment efficacy. Methodologically, researchers analyze drug sensitivity using databases like CellMiner and perform in vitro drug sensitivity assays with THBS1-manipulated cancer cell lines to establish causative relationships .
THBS1 regulates intestinal mucosal wound repair through several molecular mechanisms. It modulates actin cytoskeletal dynamics and focal adhesion formation in intestinal epithelial cells in a TGF-β1-dependent manner . Specifically, THBS1 suppresses RhoA/myosin light chain (MLC) contractility while enhancing Rac1 signaling in migrating epithelial cell sheets . Experimental approaches to study these mechanisms include in vivo biopsy-induced colonic mucosal wound repair assays, bone marrow chimera experiments to distinguish cell-type-specific contributions, and in vitro 2D primary cultures of colonic epithelial cells to analyze migration dynamics .
During tissue injury and repair, THBS1 expression is dynamically regulated. In colonic mucosal injuries, qPCR analysis shows that THBS1 mRNA is significantly upregulated following biopsy-induced wounds, with expression peaking at specific time points during the healing process . In situ hybridization using RNAscope technology has revealed that THBS1 expression forms a gradient within healing wounds, with highest expression at the leading edge of migrating epithelial cells . This spatiotemporal regulation is critical for coordinating cell migration during wound healing. Experimental approaches to study this include time-course analyses of THBS1 expression at wound sites and laser capture microdissection to isolate specific regions of healing tissue.
THBS1 is secreted in response to inflammation and has been implicated in tissue injury, inflammatory diseases, and fibrosis . In acute-on-chronic liver failure (ACLF), THBS1 acts as a disease development-related biomarker that promotes inflammatory responses and hepatocellular apoptosis . Research methodologies for studying THBS1 in inflammation include transcriptome analyses of peripheral blood mononuclear cells (PBMCs), quantitative PCR validation, and enzyme-linked immunosorbent assays (ELISA) to measure plasma THBS1 levels in relation to inflammatory markers .
Several experimental models have proven valuable for investigating THBS1 in inflammatory contexts. These include ACLF rat models and hepatocyte-specific THBS1 knockout mice for studying liver inflammation . For intestinal inflammation, researchers have utilized biopsy-induced colonic injury models in THBS1-null mice and dextran sodium sulfate (DSS)-induced colitis models . These approaches allow researchers to assess THBS1's functional contributions through both loss-of-function and gain-of-function experiments. Importantly, experiments with bone marrow chimeras derived from THBS1-deficient mice have demonstrated that both hematopoietic- and non-hematopoietic-derived THBS1 significantly contribute to tissue repair processes .
Advanced techniques for analyzing THBS1's spatiotemporal expression include RNA sequencing of laser capture micro-dissected tissue regions, which has been used to demonstrate that THBS1 is one of the genes with highest connectivity at tumor borders in glioblastoma . In situ hybridization techniques such as RNAscope provide high-resolution visualization of THBS1 mRNA expression patterns in intact tissues . To capture THBS1's dynamic regulation, time-course experiments combined with high-resolution imaging of wound edges have revealed gradients of THBS1 expression that coordinate collective cell migration . For quantitative assessment across larger samples, digital pathology approaches can analyze THBS1 immunohistochemistry staining patterns in relation to clinical outcomes.
Several approaches exist for targeting THBS1 function in experimental systems. These include genetic manipulation through THBS1 silencing or knockout models, such as the hepatocyte-specific THBS1 knockout mice that showed improved survival, repressed inflammatory cytokines, enhanced anti-inflammatory mediators, and reduced hepatocellular apoptosis in liver injury models . For more targeted intervention, researchers have used specific inhibition of THBS1/CD47 interaction using antagonist peptides to decrease cell invasion in glioblastoma models . Antibody-based approaches include direct injection of anti-THBS1 antibodies into preovulatory follicles, which reduced follicle rupture and oocyte release in primates, demonstrating THBS1's critical role in follicular angiogenesis . Each approach offers distinct advantages for studying different aspects of THBS1 biology.
THBS1 has shown significant potential as a prognostic biomarker in several diseases. In acute-on-chronic liver failure, plasma THBS1 demonstrated good predictive ability for short-term mortality, with areas under the receiver operating characteristic curve (AUROC) of 0.7445 and 0.7175 at 28 and 90 days, respectively . ACLF patients with high-risk short-term mortality were identified based on a plasma THBS1 optimal cut-off value (< 28 μg/ml) . In cancer research, THBS1 overexpression in gastric cancer was associated with poor prognosis and identified as an independent risk factor . Methodologically, validating THBS1 as a biomarker requires multiple approaches including transcriptome analyses, protein quantification in patient samples, and correlation with clinical outcomes across large, diverse patient cohorts.
Therapeutic strategies targeting THBS1 or its pathways are emerging as promising approaches in various diseases. In cancer, inhibiting THBS1 function has been shown to reduce tumor cell invasion and growth, both alone and in combination with anti-angiogenic therapy . Specific inhibition of the THBS1/CD47 interaction using antagonist peptides has demonstrated efficacy in decreasing cell invasion . For inflammatory conditions, targeting THBS1 may help modulate excessive inflammatory responses, as suggested by improved outcomes in hepatocyte-specific THBS1 knockout mice . Development of effective THBS1-targeted therapies requires careful consideration of its context-dependent functions and potential off-target effects, necessitating rigorous preclinical testing across multiple disease models.
Thrombospondin-1 is a large protein with a molecular mass ranging from 150 to 180 kDa. It is secreted as a disulfide-linked homotrimer, meaning it consists of three identical subunits linked by disulfide bonds . The protein contains several distinct domains, including:
Thrombospondin-1 is known for its role in modulating cell adhesion, motility, and growth. It interacts with a variety of cell adhesion receptors, including CD36, αv integrins, β1 integrins, syndecan, and integrin-associated protein (IAP or CD47) . Additionally, it interacts with numerous proteases involved in angiogenesis, such as plasminogen, urokinase, matrix metalloproteinase, thrombin, cathepsin, and elastase .
One of the key functions of thrombospondin-1 is its ability to inhibit neovascularization and tumorigenesis in healthy tissue. It has been shown to both positively and negatively modulate endothelial cell adhesion, motility, and growth . This dual role makes it a potential target for therapeutic applications in cancer treatment.
Recombinant human thrombospondin-1 is produced using various expression systems, including mouse myeloma cell lines and human embryonic kidney (HEK) 293 cells . The recombinant protein is typically purified to high levels of purity and is used in various research applications, including studies on cell migration, proliferation, and angiogenesis.
Recombinant thrombospondin-1 is often used in functional assays to study its binding ability and interactions with other proteins. For example, it has been shown to bind to recombinant human vascular endothelial growth factor (VEGF) in a functional ELISA .
Due to its role in modulating cell adhesion, motility, and growth, thrombospondin-1 has potential therapeutic applications in various fields, including cancer therapy, wound healing, and tissue engineering. Recombinant versions of thrombospondin-1 and its functional domains are being explored for their ability to inhibit tumor growth and angiogenesis .