SH3BGRL (SH3 domain-binding glutamic acid-rich-like protein) is a 114-amino acid protein encoded by the SH3BGRL gene located on chromosome Xq13.3. It belongs to the SH3BGR family and is characterized by a proline-rich sequence (PLPPQIF) containing SH3-binding (PXXP) and Homer EVH1-binding (PPXXF) motifs . SH3BGRL functions as an adapter protein, mediating interactions between proteins or proteins and mRNAs, and is implicated in diverse cellular processes, including signaling, autophagy, and cancer biology .
SH3BGRL acts as a scaffold protein, regulating pathways critical for cell survival, stress response, and metabolism.
SH3BGRL modulates autophagy, a catabolic process involving lysosomal degradation of cellular components. In breast cancer, it promotes chemoresistance by:
Enhancing PIK3C3 translation: Activates autophagy initiation .
Stabilizing ATG12: Maintains autophagosome formation .
In contrast, in liver cancer, SH3BGRL suppresses tumor progression by upregulating ATG5-dependent autophagic cell death .
c-Src Activation:
Ubiquitin Ligase Interaction: Associates with translating ribosomes and may regulate protein degradation via ubiquitination .
SH3BGRL exhibits context-dependent roles in cancer, varying by tissue and mutation status.
Prognostic Value:
Therapeutic Target:
A single amino acid substitution (V108A or R76C) in human SH3BGRL restores oncogenic activity, mimicking murine SH3BGRL . This highlights the importance of mutation screening in precision oncology.
SH3BGRL is a human gene that encodes a small protein consisting of 114 amino acids. The protein contains an SH3 (Src homology 3) binding motif and shares significant homology with the SH3BGR gene, specifically 60% identity and 84% conservation at the amino acid level with the middle, proline-rich region of SH3BGR . The gene appears to be part of a larger family of proteins characterized by highly conserved proline-rich functional domains. SH3BGRL belongs to a family that includes multiple isoforms (SH3BGRL, SH3BGRL2, SH3BGRL3), which likely have related but distinct cellular functions .
The SH3BGRL gene has been mapped through fluorescent in situ hybridization to Chromosome Xq13.3 . Its location on the X chromosome is particularly significant because research has demonstrated that SH3BGRL is among the genes that escape X-inactivation in humans . This chromosomal location has important implications for expression patterns in males versus females and potential roles in X-linked disorders.
Unlike its homolog SH3BGR (which is primarily expressed in heart and skeletal muscle), the SH3BGRL gene produces a 1.9 kb transcript that has been detected in all tissues examined . This ubiquitous expression pattern suggests that SH3BGRL may perform fundamental cellular functions across different tissue types rather than having a tissue-specific role. Researchers investigating SH3BGRL should consider this broad expression pattern when designing experiments and interpreting results.
The SH3BGRL protein appears to be highly evolutionarily conserved, sharing approximately 95% identity with its mouse homologue . This remarkable conservation across species suggests that SH3BGRL likely performs essential cellular functions that have been maintained throughout mammalian evolution. The strong conservation provides researchers with the opportunity to use model organisms such as mice for studying SH3BGRL function with potential relevance to human biology.
Research methods to study this phenomenon include:
RNA-seq analysis comparing male and female expression levels
Allele-specific expression analysis in female samples
DNA methylation profiling of the SH3BGRL locus in females
The proline-rich region is a defining feature of this protein family and likely serves as an interaction interface for binding partners including SH3 domain-containing proteins. Advanced structural biology techniques that can be employed to further characterize these proteins include:
X-ray crystallography of purified protein
Cryo-electron microscopy
Nuclear magnetic resonance (NMR) spectroscopy
Computational modeling using tools like AlphaFold3
Recent research has explored potential interactions between SH3BGRL isoforms and monomeric actin. Although the primary sequence of the actin-binding loop identified in related proteins is not conserved in human SH3BGRL isoforms, computational approaches have been employed to investigate potential heterodimeric complexes .
Researchers have utilized AlphaFold3 and AlphaFold-Multimer to generate models of potential SH3BGRL/-2/-3 – G-actin complexes. These computational predictions are evaluated using predicted template modelling (pTM) and interface predicted template modelling (ipTM) scores, which assess the accuracy of the predicted folding and interaction interfaces, respectively .
Experimental approaches to validate these predictions might include:
Co-immunoprecipitation assays
Fluorescence resonance energy transfer (FRET)
Surface plasmon resonance
Actin polymerization assays in the presence of purified SH3BGRL proteins
For investigating SH3BGRL gene regulation, researchers should consider a multi-faceted approach:
Promoter analysis: Identify regulatory elements using reporter assays with constructs containing the SH3BGRL promoter region
Epigenetic profiling: Analyze DNA methylation patterns and histone modifications at the SH3BGRL locus using techniques such as bisulfite sequencing and ChIP-seq
Transcription factor binding: Employ ChIP-seq or DNA-protein interaction assays to identify transcription factors that regulate SH3BGRL expression
X-inactivation escape mechanisms: Given SH3BGRL's status as an escape gene, investigate chromatin accessibility using ATAC-seq comparing the active and inactive X chromosomes in female cells
Studies would benefit from comparing these patterns between different cell types and under various physiological conditions to understand context-dependent regulation.
Distinguishing between SH3BGRL isoforms (SH3BGRL, SH3BGRL2, SH3BGRL3) experimentally requires techniques that can detect specific protein variants:
Western blotting with isoform-specific antibodies: Develop and validate antibodies that recognize unique epitopes in each isoform
Mass spectrometry: Use targeted proteomics approaches to identify isoform-specific peptides
RT-qPCR with isoform-specific primers: Design primers targeting unique regions of each transcript variant
CRISPR-based tagging: Generate cell lines with fluorescently tagged endogenous isoforms to track localization and expression
Isoform-specific knockdown: Design siRNAs or shRNAs targeting unique regions of each isoform to study their individual functions
These approaches would enable researchers to determine the relative abundance and distinct functions of each SH3BGRL isoform in different cellular contexts.
Current research employs sophisticated computational approaches to predict SH3BGRL interactions:
AlphaFold3 and AlphaFold-Multimer: These tools have been used to generate models of potential SH3BGRL-actin complexes . The reliability of these predictions is assessed using pTM (predicted template modeling) and ipTM (interface predicted template modeling) scores, which evaluate the accuracy of predicted protein folding and interaction interfaces, respectively.
Molecular dynamics simulations: These can be used to model the dynamic behavior of predicted protein complexes over time, providing insights into the stability and conformational changes of the interactions.
Protein-protein docking: Tools like HADDOCK, ClusPro, or Rosetta can predict binding modes between SH3BGRL and potential partner proteins based on structural information.
Interaction network analysis: Integration of proteomics data, literature-based evidence, and predictive algorithms can help construct comprehensive interaction networks for SH3BGRL.
For optimal results, researchers should combine multiple computational approaches and validate predictions with experimental evidence.
SH3BGRL's classification as an X-inactivation escape gene has significant implications for disease research :
Dosage effects: Escape from X-inactivation may result in higher expression levels in females compared to males, potentially contributing to sex-biased disease manifestations.
Contribution to polyX phenotypes: Research has shown an excess of escaping genes associated with mental retardation consistent with common phenotypes in polyX karyotypes . As an escape gene, SH3BGRL may contribute to neurological phenotypes in individuals with X chromosome aneuploidies.
Population differences: Evidence indicates differences between populations in the propensity to permit escape from X-inactivation . This could result in population-specific disease risks or manifestations related to SH3BGRL function.
Individual variation: Studies have identified both "hyper-escapee" and "hypo-escapee" females in the human population, with significant differences in their propensity to allow genes to escape inactivation . This individual variation could contribute to phenotypic heterogeneity in conditions involving SH3BGRL dysfunction.
Methodologically, researchers should account for these factors by:
Including sex as a biological variable in study design
Analyzing population-specific expression patterns
Assessing individual variation in SH3BGRL expression in clinical cohorts
Correlating escape status with phenotypic manifestations
To validate computational predictions about SH3BGRL structure and interactions, researchers should employ a strategic experimental approach:
This comprehensive validation strategy ensures that computational predictions translate to biologically relevant insights about SH3BGRL function.
Several significant challenges complicate the study of SH3BGRL function:
Functional redundancy: The existence of multiple SH3BGRL family members (SH3BGRL, SH3BGRL2, SH3BGRL3) suggests potential functional redundancy, making it difficult to attribute specific cellular roles to individual isoforms.
Ubiquitous expression: The widespread expression of SH3BGRL across tissues complicates the identification of tissue-specific functions and may mask important specialized roles.
X-inactivation escape complexity: The variable escape from X-inactivation observed between individuals and populations introduces heterogeneity that must be accounted for in experimental design and data interpretation.
Limited functional data: Despite structural information and interaction predictions, there remains a gap in understanding the precise biochemical and cellular functions of SH3BGRL proteins.
Protein-protein interaction verification: While computational approaches predict potential interactions , experimental validation of these interactions in physiologically relevant contexts remains challenging.
Addressing these challenges will require integrated approaches combining genomics, proteomics, structural biology, and functional studies in appropriate cellular models.
As a gene that escapes X-inactivation , SH3BGRL may contribute to sex-specific differences in gene expression through several mechanisms:
Dosage effects: Higher expression in females due to transcription from both X chromosomes could directly impact cellular processes involving SH3BGRL.
Regulatory network influences: If SH3BGRL functions in gene regulation pathways, differential expression between sexes could propagate to affect downstream genes, amplifying sex-biased expression patterns.
Individual variability: The documented existence of "hyper-escapee" and "hypo-escapee" females suggests that SH3BGRL may contribute to phenotypic variability among females in addition to male-female differences.
Tissue-specific escape patterns: X-inactivation escape can vary between tissues, potentially leading to tissue-specific sex differences in SH3BGRL expression.
Research approaches to investigate these effects include:
Single-cell RNA-seq comparing male and female cells
Analysis of SH3BGRL expression correlation with other escape genes
Tissue-specific analysis of escape patterns
Cellular models with modulated SH3BGRL expression to assess effects on global gene expression
The SH3 Domain Binding Glutamic Acid-Rich Protein Like (SH3BGRL) is a member of a small family of proteins that are characterized by their ability to bind to SH3 domains. These proteins are involved in various cellular processes, including signal transduction, cytoskeletal organization, and cellular migration. The human recombinant form of SH3BGRL has been studied for its potential roles in cancer and other diseases.
The SH3BGRL gene is located on human chromosome 1 at the position 1p36.11 . The gene encodes a protein that is approximately 10.5 kDa in size and has an isoelectric point of 5.0 . The SH3BGRL protein family includes several homologous proteins, such as SH3BGR, SH3BGRL, and SH3BGRL3, which are located on different chromosomes and have varying expression patterns .
SH3BGRL is ubiquitously expressed in human tissues, with higher expression levels observed in certain tissues such as the spleen, thymus, and various arteries . The protein is found in multiple cellular compartments, including the nucleus, cytoplasm, and extracellular exosomes . This widespread expression suggests that SH3BGRL may play a fundamental role in various cellular functions.
While the precise functions of SH3BGRL are not fully understood, it is known to be involved in redox homeostasis and electron transport chain processes . The protein shows significant similarity to glutaredoxin 1 of E. coli, which is an oxidoreductase involved in reducing intracellular protein disulfides . However, SH3BGRL lacks the enzymatic activity of glutaredoxins and may instead act as a regulator of redox activity .
SH3BGRL has been implicated in cancer, particularly in glioblastoma multiforme, where it is up-regulated compared to normal cerebral tissue . The protein has also been found to be overexpressed in several other tumors, suggesting a potential role in tumorigenesis . Recent studies have indicated that SH3BGRL may interact with members of the EGFR family, which are known to be involved in cancer progression .
SH3BGRL interacts with myosin 1c in a calcium-dependent manner, which modulates cellular migration . This interaction is crucial for the regulation of cytoskeletal dynamics and cell motility. The binding of SH3BGRL to myosin 1c requires the presence of calcium, suggesting that calcium-driven mechanisms may regulate the conformation and activity of myosin 1c .