SH3BGRL2 is aberrantly expressed in multiple cancers:
In breast cancer, SH3BGRL2 mRNA and protein levels are reduced in 67–93% of primary tumors compared to normal tissues . Similarly, 62.5% of ccRCC tumors show decreased SH3BGRL2 expression .
Growth suppression: Overexpression inhibits proliferation and colony formation (in vitro) and tumorigenesis (in vivo) .
Metastasis promotion: Enhances cell migration, invasion, and lung metastasis via transcriptional repression of SPTAN1 and SPTBN1 (cytoskeletal regulators) .
Regulation: TGF-β1 activates SH3BGRL2 through the Smad pathway, linking it to metastasis .
SH3BGRL2 loss drives proliferation, migration, and invasion by activating the Hippo/TEAD1-Twist1 pathway, promoting epithelial-mesenchymal transition (EMT) .
Key mechanism: SH3BGRL2 downregulation elevates LATS1/2 expression, leading to YAP phosphorylation, nuclear translocation, and Twist1-mediated EMT .
SH3BGRL2 binds SPTAN1 and SPTBN1, reducing their expression and destabilizing the cytoskeleton to facilitate metastasis in breast cancer .
In breast cancer, SH3BGRL2 is transcriptionally activated by TGF-β1/Smad signaling .
In ccRCC, SH3BGRL2 loss enables TEAD1 to bind the Twist1 promoter, upregulating this EMT master regulator .
Prognostic marker: Low SH3BGRL2 correlates with advanced TNM stage, recurrence, and poor survival in ccRCC (HR = 0.329, p < 0.05) .
Therapeutic target: Restoring SH3BGRL2 expression could inhibit metastasis in breast cancer or ccRCC by modulating Hippo or TGF-β pathways .
Regulatory mechanisms: Unknown whether SH3BGRL2 downregulation in cancers results from methylation, miRNA regulation, or mutations .
Pathway conservation: Does SH3BGRL2-Hippo/TEAD1-Twist1 signaling apply to other cancers (e.g., lung, ovarian)? .
Functional variability: Why does SH3BGRL2 act as a metastasis promoter in breast cancer but a suppressor in ccRCC?
SH3BGRL2 (SH3 domain-binding glutamic acid-rich-like protein 2) belongs to the SH3BGR gene family, which includes SH3BGR, SH3BGRL, SH3BGRL2, and SH3BGRL3. It contains a highly conserved proline-rich domain that facilitates interactions with proteins containing specific binding modules such as Src homology 3 (SH3), WW, and Enabled/VASP homology-1 (EVH1) domains. SH3BGRL2 is located in the nucleus and perinuclear region, containing both SH3 and EVH1 domains which play central roles in cell growth, adhesion, and migration . This protein family shows varying expression patterns across different developmental stages and organ types, suggesting their dysregulation may contribute to various pathological conditions .
While SH3BGRL2 remains relatively understudied compared to other proteins, available evidence indicates its involvement in several normal physiological processes. Research suggests SH3BGRL2 plays crucial roles in erythroid differentiation, tissue development, and may be implicated in metabolic conditions like diabetes . Specifically, Tong et al. demonstrated SH3BGRL2's importance in nervous system development and intestine formation in zebrafish models . The protein's structural domains (SH3 and EVH1) are associated with cytoskeletal organization, cell adhesion, and signaling pathway regulation, which are fundamental to maintaining cellular homeostasis in normal tissues .
The regulation of SH3BGRL2 involves multiple mechanisms, with growth factors and cytokines playing significant roles. Research has identified that transforming growth factor-β1 (TGF-β1) transcriptionally activates SH3BGRL2 expression through the canonical TGF-β receptor-Smad pathway in breast cancer cells . This regulatory relationship is particularly significant given TGF-β1's established role in tumor progression and metastasis. Additionally, bioinformatic analyses from TCGA and GEO databases suggest tissue-specific regulation, as SH3BGRL2 expression varies considerably across different tissue types and shows altered patterns in malignant versus normal tissues . The complex transcriptional control of SH3BGRL2 likely reflects its context-dependent functions in different cellular environments.
The loss of SH3BGRL2 in ccRCC initiates a complex signaling cascade through the Hippo/TEAD1/Twist1 pathway that promotes epithelial-mesenchymal transition (EMT) and subsequent tumor progression. Mechanistically, this process involves multiple sequential steps: (i) loss of SH3BGRL2 induces increased expression of LATS1/2, leading to YAP phosphorylation, activation, and nuclear translocation; (ii) once in the nucleus, activated YAP binds to its co-transcriptional factor TEAD1; and (iii) TEAD1 directly binds to the Twist1 promoter, enhancing its expression and ultimately inducing EMT . EMT is a critical process that enables cancer cells to acquire migratory and invasive properties, facilitating tumor growth and metastasis. This mechanism provides valuable insights into how SH3BGRL2 downregulation contributes to the aggressive phenotype observed in high-grade ccRCC and offers potential targets for therapeutic intervention along this signaling axis .
In breast cancer, SH3BGRL2 exerts its pro-metastatic effects primarily through interaction with cytoskeletal proteins. Research has revealed that SH3BGRL2 interacts with and transcriptionally represses spectrin alpha, non-erythrocytic 1 (SPTAN1) and spectrin beta, non-erythrocytic 1 (SPTBN1), which are essential cytoskeletal proteins . Functional rescue assays demonstrated that depletion of SH3BGRL2 reduced breast cancer cell invasive potential, which could be partially rescued by knockdown of SPTAN1 and SPTBN1 . This interaction with spectrin proteins provides a mechanistic explanation for SH3BGRL2's pro-metastatic effects, as alterations in cytoskeletal organization are fundamental to cancer cell migration and invasion. The SH3 and EVH1 domains in SH3BGRL2 likely facilitate these protein-protein interactions, enabling modulation of cell adhesion and migration machinery . Understanding these molecular mechanisms could inform the development of targeted therapies to disrupt SH3BGRL2-mediated cytoskeletal reorganization and potentially inhibit metastasis.
Comprehensive investigation of SH3BGRL2 requires multiple complementary experimental approaches. For expression analysis, researchers should employ RT-PCR, western blotting, and immunohistochemistry to examine both mRNA and protein levels in patient samples and cell lines . Gain- and loss-of-function studies using overexpression vectors and RNA interference (siRNA/shRNA) or CRISPR-Cas9 systems are essential for evaluating SH3BGRL2's functional roles in cellular processes like proliferation, migration, and invasion . In vitro assays should be complemented with in vivo xenograft models to assess effects on tumor growth and metastasis, as demonstrated in studies using both traditional cell line-derived xenografts and patient-derived xenograft (PDX) models . For mechanistic investigations, chromatin immunoprecipitation (ChIP) assays and luciferase reporter assays have proven valuable for elucidating SH3BGRL2's involvement in transcriptional regulation, while co-immunoprecipitation experiments can identify protein-protein interactions . Additionally, bioinformatic analyses of publicly available databases (TCGA, GEO) provide valuable insights into clinical correlations and potential signaling pathways for further investigation .
Accurate assessment of SH3BGRL2 in clinical samples presents several technical challenges that researchers must address. First, the selection of appropriate antibodies with high specificity and sensitivity is crucial, as cross-reactivity with other SH3BGR family members could lead to misinterpretation of results . Second, researchers must carefully consider the heterogeneity of tumor tissues, as SH3BGRL2 expression varies within tumors and between patients; this necessitates adequate sampling and potentially microdissection techniques to isolate specific cell populations . Third, determining the optimal cut-off values for categorizing "high" versus "low" SH3BGRL2 expression requires robust statistical approaches, such as receiver operating characteristic (ROC) curve analysis as employed in previous studies . Fourth, correlation of expression levels with clinical outcomes requires comprehensive patient data and appropriate statistical models that account for confounding variables . Finally, since SH3BGRL2 can have context-dependent functions (as seen in its dual role in breast cancer), researchers must evaluate not only expression levels but also activation status and interaction partners to fully understand its clinical implications .
Given SH3BGRL2's context-dependent functions, researchers must employ systematic approaches to characterize its diverse roles across different cellular environments. First, comprehensive phenotypic profiling using a panel of functional assays (proliferation, colony formation, migration, invasion, EMT markers) in multiple cell types is essential to capture the full spectrum of SH3BGRL2's effects . Second, domain-specific mutational analysis can help identify which structural features (SH3, EVH1, or proline-rich domains) mediate specific functions in different contexts . Third, interactome analysis using techniques like mass spectrometry-based proteomics following immunoprecipitation can reveal cell type-specific binding partners and signaling networks . Fourth, pathway analysis using pharmacological inhibitors or genetic manipulation of suspected signaling mediators (e.g., Hippo pathway components in ccRCC, spectrin proteins in breast cancer) can elucidate the molecular mechanisms underlying context-specific functions . Finally, in vivo models that recapitulate tissue-specific microenvironments, such as orthotopic xenografts or genetically engineered mouse models, provide physiologically relevant systems to validate in vitro findings and explore the impact of the tumor microenvironment on SH3BGRL2 function .
Therapeutic approaches targeting SH3BGRL2 would need to account for its context-dependent functions across different cancer types. For cancers where SH3BGRL2 acts primarily as a tumor suppressor (e.g., ccRCC), strategies to restore or enhance its expression might be beneficial. This could include epigenetic modifiers to reverse potential silencing mechanisms or small molecules that mimic SH3BGRL2's tumor-suppressive functions . Alternatively, targeting downstream effectors in the Hippo/TEAD1/Twist1 signaling pathway, such as YAP inhibitors or TEAD-YAP interaction disruptors, could counteract the effects of SH3BGRL2 loss . For cancers where SH3BGRL2 exhibits dual functions (e.g., breast cancer), more nuanced approaches are needed. While inhibiting SH3BGRL2 might reduce metastasis, it could potentially promote tumor growth . Therefore, combination therapies targeting both SH3BGRL2 and complementary pathways might be necessary. Additionally, since TGF-β1 regulates SH3BGRL2 expression, TGF-β pathway inhibitors could indirectly modulate SH3BGRL2 levels . As research progresses, personalized therapeutic strategies based on individual patient's SH3BGRL2 expression profiles and cancer type could optimize treatment outcomes.
SH3BGRL2 expression patterns could significantly influence clinical decision-making and trial design. For ccRCC patients, where approximately 40% develop metastases after surgical removal of the primary tumor, SH3BGRL2 status could identify high-risk patients who might benefit from adjuvant therapy . Low SH3BGRL2 expression, as an independent prognostic factor for disease-free survival, could serve as a selection criterion for more aggressive treatment regimens or closer surveillance protocols . In clinical trials, stratifying patients based on SH3BGRL2 expression could help identify subgroups most likely to respond to specific interventions, particularly those targeting the Hippo/TEAD1/Twist1 pathway . For breast cancer patients, the dual role of SH3BGRL2 suggests that its expression patterns might inform different treatment strategies for preventing primary tumor growth versus metastatic spread . Additionally, since SH3BGRL2 is regulated by TGF-β1, its expression might predict responsiveness to TGF-β pathway inhibitors . As more data accumulates, incorporating SH3BGRL2 assessment into molecular profiling panels could enhance personalized medicine approaches, guiding treatment selection and sequence to optimize outcomes for individual patients.
When faced with contradictory findings regarding SH3BGRL2 function, researchers should systematically evaluate several factors that might explain these discrepancies. First, consider biological context differences: SH3BGRL2 exhibits tissue-specific functions, as evidenced by its tumor-suppressive role in ccRCC versus its dual function in breast cancer . Second, examine methodological variations: differences in experimental techniques, cell lines, animal models, and analytical approaches can significantly impact results. Third, assess the temporal dimension: SH3BGRL2's effects may vary depending on disease stage or cellular state, with potentially different roles during initial tumor formation versus progression and metastasis . Fourth, evaluate interaction partner availability: since SH3BGRL2 functions through protein-protein interactions, the presence or absence of specific binding partners in different experimental systems could alter its activity . Fifth, consider post-translational modifications: unidentified modifications might modulate SH3BGRL2 function in context-dependent ways. When publishing findings, researchers should clearly describe these contextual factors and avoid overgeneralizing results across cancer types or experimental systems without adequate validation.
SH3 Domain Binding Glutamic Acid-Rich Protein Like 2 (SH3BGRL2) is a protein encoded by the SH3BGRL2 gene in humans. This protein is part of a family of proteins known for their SH3 domain-binding capabilities and glutamic acid-rich sequences. These proteins play significant roles in various cellular processes, including signal transduction and cytoskeletal organization.
The SH3BGRL2 gene is located on human chromosome 1. The protein encoded by this gene has a thioredoxin-like fold, which is a common structural motif involved in redox reactions. However, unlike typical thioredoxin proteins, SH3BGRL2 lacks the cysteines necessary for catalytic activity . This suggests that while it shares structural similarities with thioredoxins, its function may differ significantly.
SH3BGRL2 is characterized by the presence of a C-terminal proline-rich sequence with SH3 domain- and HOMER EVH1 domain-binding motifs . These motifs are crucial for protein-protein interactions, indicating that SH3BGRL2 may play a role in assembling protein complexes and facilitating intracellular signaling pathways.
Although the precise biological functions of SH3BGRL2 are still under investigation, it is believed to be involved in regulating redox activity within cells. This regulatory role is essential for maintaining cellular homeostasis and protecting cells from oxidative stress. Additionally, proteins with similar structures, such as glutaredoxins and thioredoxins, are known to be upregulated in various cancers, suggesting a potential link between SH3BGRL2 and cancer biology .
Research into SH3BGRL2 is ongoing, with studies focusing on its role in cancer and other diseases. Understanding the function and regulation of this protein could provide insights into new therapeutic targets for treating conditions associated with oxidative stress and abnormal cell signaling.