FAM84A is a 315-amino acid protein with a molecular weight of 34.9 kDa, belonging to the FAM84 protein family . Key features include:
The protein shows cytoplasmic localization in hepatocytes and cancer cells, with expression primarily detected in testis under normal physiological conditions .
FAM84A exhibits oncogenic properties through multiple pathways:
Phosphorylation at Serine 38 correlates with morphological changes in colon cancer cells, though this modification is absent in liver tumors . In papillary thyroid carcinoma (PTC), FAM84A knockdown reduces tumor size by 58% in xenograft models and decreases Ki-67 proliferation markers by 42% .
FAM84A overexpression shows diagnostic and prognostic value across malignancies:
In PTC, high FAM84A expression correlates with:
82% sensitivity for lymph node metastasis
2.4-fold increased risk of recurrence
FAM84A is modulated by multiple upstream factors:
The miR-874-3p/FAM84A axis demonstrates clinical relevance, with inverse correlation (r=-0.68, p<0.001) observed in 80 PTC specimens .
Key experimental approaches in FAM84A studies:
Current research priorities include:
Developing small-molecule inhibitors targeting FAM84A's collagen-like domain
Exploring combination therapies with Wnt/β-catenin pathway inhibitors
Validating FAM84A as a liquid biopsy marker in pancreatic and bladder cancers
Investigating isoform-specific functions through alternative splicing analysis
FAM84A is a protein-coding gene that has been characterized as a novel collagen-type protein based on its amino acid sequence identity with human collagen type X and α1. The protein contains several functional domains, including potential phosphorylation sites that may be critical for its activity. Notably, serine 38 has been identified as a potentially important residue, as phosphorylation at this site has been associated with morphological changes in cells . Immunohistochemistry analyses have revealed that FAM84A protein exhibits predominantly cytoplasmic localization in both normal and cancerous tissues . This cytoplasmic distribution suggests that FAM84A may interact with cytoplasmic signaling components or structural elements involved in cellular migration and proliferation.
FAM84A expression is tightly regulated through multiple mechanisms that vary by tissue type and physiological context. At the transcriptional level, the nuclear xenobiotic receptor CAR (Constitutive Androstane Receptor) has been identified as a key regulator of FAM84A expression, capable of activating the FAM84A promoter as demonstrated in HepG2 cell-based reporter assays . Interestingly, FAM84A regulation shows sex-dependent patterns, with higher basal expression levels observed in female mice compared to males, though phenobarbital (PB) treatment induces expression only in males . This regulation also exhibits strain-dependency, with PB treatment significantly inducing FAM84A mRNA in C3He male mice but not in C57BL6 male mice . At the post-transcriptional level, miR-874-3p acts as a negative regulator of FAM84A expression . In pathological states, particularly cancer, FAM84A is frequently upregulated, with expression levels correlating with clinical parameters such as tumor size and advanced disease stages .
Researchers have successfully employed several complementary techniques to study FAM84A expression. For RNA detection, real-time PCR (qRT-PCR) remains the gold standard, using specific primers for FAM84A mRNA quantification normalized to housekeeping genes like GAPDH . The forward and reverse primers for mouse FAM84A cDNA that have been validated include: 5′-CCTCGCCGCGTCATTG-3′ and 5′-CAGTGGGCAACTCGCTGTAG-3′ . For human FAM84A, primers such as 5'‐ATTCGGCTCGGGGTAGAG‐3' and 5'‐TCTTCCTCATCATCCGAGAA‐3' have been used effectively . At the protein level, Western blotting provides quantitative assessment using commercially available antibodies, such as those from Abcam (used at 1:1500 dilution) . Standard SDS-PAGE followed by transfer to PVDF membranes with 5% nonfat milk blocking has proven effective . Immunohistochemistry (IHC) enables visualization of FAM84A localization in tissue samples and has been instrumental in determining its cytoplasmic distribution pattern . Each method offers distinct advantages, and combining multiple approaches provides more robust validation of expression patterns.
While detailed three-dimensional crystal structures for FAM84A have not been extensively reported in the literature, functional studies have identified several important structural features. FAM84A shares sequence similarity with collagen proteins, which suggests potential structural similarities and related functions . The protein contains specific phosphorylation sites, with serine 38 in mouse FAM84A identified as particularly significant. Phosphorylation at this residue has been associated with morphological changes in cells, although interestingly, in liver and liver tumors, FAM84A was not found to be phosphorylated at this position . This observation suggests tissue-specific regulation of post-translational modifications that may influence FAM84A function. Site-directed mutagenesis approaches have been employed to generate serine 38 to alanine mutations using the Quickchange site-directed mutagenesis kit and specific primers (5′-GGTTGCCTACTTCTTCGCGGATGAGGAGGAGGA-3′ and 5′-TCCTCCTCCTCATCCGCGAAGAAGTAGGCAACC-3′), enabling functional studies of this residue .
The expression pattern of FAM84A varies significantly across different tissue types under normal physiological conditions. In mouse models, FAM84A shows relatively low expression in the liver under basal conditions, but expression can be induced by treatments such as phenobarbital in a sex-dependent and strain-dependent manner . Interestingly, the basal level of FAM84A mRNA has been observed to be higher in female mice compared to males, suggesting hormonal regulation may play a role in its expression . FAM84A has been detected in brain tissue, which has been utilized as a source for cDNA amplification due to its higher expression levels compared to liver tissue . Within the liver, immunohistochemistry has shown that FAM84A protein expression is particularly prominent in hepatocytes around the central vein . This heterogeneous distribution within tissues suggests cell-type specific regulation and potentially specialized functions in different cellular contexts.
FAM84A plays a significant role in promoting cancer cell proliferation and survival through multiple mechanisms. In papillary thyroid carcinoma (PTC), knockdown of FAM84A significantly reduced colony formation ability and attenuated cell growth as demonstrated by CCK-8 assays and EdU incorporation assays, which measure DNA synthesis . Flow cytometric analysis revealed that FAM84A knockdown induced cell cycle arrest at the G0/G1 phase and increased apoptotic rates in PTC cell lines . At the molecular level, FAM84A influences the expression of cell cycle regulators, with its downregulation leading to reduced levels of CDK4 and CDK6, which are critical for G1/S phase transition . Additionally, FAM84A affects apoptotic pathways, with its knockdown decreasing expression of the anti-apoptotic protein Bcl-2 while increasing levels of the pro-apoptotic protein Bax . The positive correlation observed between FAM84A expression and Ki-67, a proliferative cell-associated antigen, further supports its role in promoting cancer cell proliferation .
FAM84A has been identified as a significant regulator of epithelial-mesenchymal transition (EMT), a critical process in cancer progression that enables tumor cells to acquire migratory and invasive properties. Studies in papillary thyroid carcinoma have demonstrated that downregulation of FAM84A inhibits PTC development by repressing EMT . Western blot analyses have revealed that FAM84A modulates the expression of key EMT markers, including E-cadherin (an epithelial marker), N-cadherin, and vimentin (mesenchymal markers) . This shift in marker expression reflects a change in cellular phenotype from an epithelial to a more mesenchymal state, which is associated with increased migratory and invasive capabilities. Additionally, the only study investigating FAM84A's biological function in colon cancer found that ectopic overexpression of FAM84A altered cell morphology and increased cell migration, suggesting its role as a pro-tumor factor through EMT promotion . These findings collectively indicate that FAM84A promotes cancer progression in part by facilitating EMT, which contributes to metastatic potential.
FAM84A has been implicated in the activation of the Wnt/β-catenin signaling pathway, a critical pathway in cancer development and progression. Studies in papillary thyroid carcinoma have demonstrated that FAM84A promotes PTC cell proliferation and metastasis by activating this pathway . Analysis of TCGA database revealed a positive correlation between FAM84A expression and β-catenin levels, supporting a functional relationship between FAM84A and Wnt/β-catenin signaling . Western blot analysis has shown that FAM84A knockdown affects β-catenin expression levels, although the precise molecular mechanisms through which FAM84A activates or interacts with this pathway remain to be fully elucidated . Potential mechanisms could include direct protein-protein interactions with components of the pathway, indirect regulation through modulation of upstream regulators, or effects on β-catenin nuclear translocation or stability. The activation of the Wnt/β-catenin pathway by FAM84A likely contributes to its observed effects on cell proliferation, survival, and EMT, as this pathway regulates numerous genes involved in these processes.
FAM84A expression alterations have been documented in multiple cancer types, with significant clinical correlations. In papillary thyroid carcinoma (PTC), analysis of TCGA and GSE66783 databases, as well as direct experimental validation, has demonstrated significantly higher FAM84A expression in PTC tissues compared to normal thyroid tissues . Clinical correlation analyses revealed that PTC patients with higher FAM84A expression tended to have larger tumor size, higher lymph node metastasis rate, and advanced TNM stage . In liver cancer models, FAM84A mRNA was found to be induced in mouse liver during phenobarbital promotion of liver tumors, with continued induction in both non-tumor and tumor tissues of tumor-bearing liver . FAM84A has also been reported to be overexpressed in various colon cancer cell lines and in many human colon tumor tissues, although not universally in all samples . These findings collectively suggest that FAM84A upregulation may be a common feature across multiple cancer types and that its expression levels may serve as a potential biomarker for disease progression and prognosis.
MicroRNAs (miRNAs) play crucial roles in post-transcriptional regulation of gene expression, and research has identified a specific relationship between miR-874-3p and FAM84A in cancer. Studies have demonstrated that miR-874-3p acts as a tumor suppressor and negatively regulates FAM84A expression . This regulatory relationship suggests a mechanism by which loss of miR-874-3p expression in cancer could contribute to elevated FAM84A levels, promoting tumor development and progression. The identification of miR-874-3p as a negative regulator of FAM84A provides important insights into the regulatory network controlling FAM84A expression in cancer cells. This relationship also suggests potential therapeutic strategies, as restoring miR-874-3p expression or function could potentially downregulate FAM84A expression and inhibit its oncogenic effects. Understanding the miR-874-3p/FAM84A axis represents an important advance in elucidating the molecular mechanisms underlying FAM84A dysregulation in cancer and offers new avenues for targeted intervention.
Several genetic manipulation strategies have proven effective for studying FAM84A function. For transient knockdown, short interfering RNA (siRNA) targeting FAM84A (si-FAM84A) has been successfully used in cell lines like TPC-1 and K-1, with transfection facilitated by Lipofectamine 3000 reagent according to manufacturer's protocols . For stable knockdown models, short hairpin RNA (shRNA) targeting FAM84A (sh-FAM84A) provides longer-term expression reduction, with stable cell lines typically selected using puromycin (3 μg/mL) for 10-15 days . These knockdown approaches effectively reduced FAM84A expression at both mRNA and protein levels, enabling studies of its function in cell proliferation, apoptosis, and other processes . For overexpression studies, full-length cDNA of FAM84A can be amplified using specific primers (such as 5′-ACCATGGGCAACCAACTGGA-3′ and 5′-GCTACTCCTTGTCGTCCACA-3′ for mouse FAM84A) and cloned into expression vectors like pcDNA3.1-His-V5-TOPO . Site-directed mutagenesis techniques can be employed to create specific mutations, such as serine 38 to alanine, for studying the functional significance of particular residues .
Multiple in vitro assays have successfully demonstrated FAM84A's functional effects on cancer-related phenotypes. For proliferation assessment, colony formation assays effectively measure clonogenic ability, while Cell Counting Kit-8 (CCK-8) assays provide quantitative measurements of cell viability and proliferation rates . The EdU (5-ethynyl-2'-deoxyuridine) incorporation assay offers precise evaluation of DNA synthesis and cell proliferation through fluorescent labeling of newly synthesized DNA . Cell cycle distribution can be analyzed by flow cytometry with propidium iodide staining, which has revealed G0/G1 phase arrest following FAM84A knockdown . Apoptosis can be quantified using Annexin V/PI staining and flow cytometry, complemented by Western blot analysis of apoptosis-related proteins like Bcl-2 and Bax . For assessing EMT and related phenotypes, Western blotting for EMT markers (E-cadherin, N-cadherin, vimentin) provides molecular evidence of phenotypic transitions . Migration and invasion capabilities can be evaluated using Transwell assays or wound healing assays. Wnt/β-catenin pathway activity can be measured through Western blot analysis of β-catenin levels or reporter assays for downstream target activation .
Animal models have provided valuable insights into FAM84A's role in cancer development and progression. Subcutaneous xenograft models using nude mice have successfully demonstrated that knockdown of FAM84A can inhibit tumor growth in vivo . In these models, cancer cells (such as TPC-1 and K-1) with stable FAM84A knockdown or control constructs are inoculated subcutaneously into the flanks of nude mice, and tumor growth is monitored over time . For liver cancer specifically, a two-step mouse model of hepatocarcinogenesis has proven informative, where tumor initiation by diethylnitrosamine (DEN) is followed by promotion with chronic phenobarbital (PB) treatment . This model has demonstrated that the nuclear receptor CAR (which regulates FAM84A) is essential for tumor development, as Car-/- mice did not develop liver tumors even after 34 weeks of PB treatment . Strain differences in susceptibility to hepatocarcinogenesis (with C3He mice being more susceptible than C57BL6 mice) correlate with differences in FAM84A induction, suggesting a potential role for FAM84A in strain-dependent cancer susceptibility . Patient-derived xenograft (PDX) models could provide additional insights by preserving tumor heterogeneity, though these have not been extensively reported for FAM84A studies.
Understanding FAM84A's protein-protein interactions is crucial for elucidating its molecular functions, though this aspect has not been extensively explored in the available literature. Several techniques could be effectively applied to this question: Co-immunoprecipitation (Co-IP) using antibodies against FAM84A followed by mass spectrometry analysis would identify proteins that physically interact with FAM84A in cellular contexts. Proximity ligation assays (PLA) could visualize and quantify interactions between FAM84A and suspected binding partners in situ. Yeast two-hybrid screening would provide an unbiased approach to identify potential interaction partners. Given FAM84A's reported effects on the Wnt/β-catenin pathway , specific Co-IP experiments targeting components of this pathway (such as β-catenin, GSK3β, and APC) would be particularly informative. For stronger validation, reciprocal Co-IPs (pulling down with antibodies against the interacting partner and blotting for FAM84A) should be performed. Tagged versions of FAM84A (with epitopes like FLAG, HA, or GFP) could facilitate these interaction studies, especially when specific FAM84A antibodies are limiting or lack sufficient specificity for certain applications.
Bioinformatic approaches offer powerful tools for generating new hypotheses about FAM84A function. Mining public databases like TCGA and GEO has already yielded important insights, revealing FAM84A upregulation in thyroid cancer compared to normal tissues . Correlation analysis between FAM84A and other genes can identify potential functional relationships, as demonstrated by the positive correlation observed between FAM84A and Ki-67 expression . Analyzing FAM84A expression across different cancer stages, grades, and molecular subtypes can provide insights into its role in disease progression. Co-expression network analysis can identify genes that are functionally related to FAM84A, while pathway enrichment analysis can reveal biological processes associated with FAM84A expression. Protein domain analysis and structural prediction tools can generate hypotheses about functional regions within the FAM84A protein. For microRNA regulation, target prediction algorithms can identify potential regulatory miRNAs beyond the already established miR-874-3p . Integration of multi-omics data (transcriptomics, proteomics, epigenomics) can provide a more comprehensive understanding of FAM84A regulation and function in different cellular contexts.
Despite progress in FAM84A research, several limitations in detection methodologies persist. Antibody specificity remains a significant challenge, with potential for cross-reactivity with related family members leading to false positive results. The relatively low expression of FAM84A in certain normal tissues, such as liver, can make detection difficult without sufficient sensitivity . This is exemplified by researchers using mouse brain cDNA for amplification due to FAM84A's higher expression there compared to liver . Post-translational modifications of FAM84A, such as phosphorylation at serine 38, may affect antibody recognition and complicate detection . The subcellular localization of FAM84A can also present challenges, as extraction methods may need to be optimized to efficiently recover cytoplasmic proteins. Additionally, distinguishing between different isoforms of FAM84A may require specific primers or antibodies. To address these limitations, researchers should validate antibodies using appropriate positive and negative controls, including FAM84A knockdown or knockout samples. Employing multiple detection methods (qRT-PCR, Western blot, IHC) can provide more robust validation of expression patterns. Developing more sensitive detection methods, such as digital PCR or highly sensitive ELISA, could improve detection in samples with low expression levels.
Contradictory findings regarding FAM84A function may arise from several factors that researchers should carefully consider. Tissue-specific context is crucial, as FAM84A regulation and function show distinct patterns across different tissues. For example, while phenobarbital induces FAM84A expression in male C3He mice, this effect is not observed in female mice or C57BL6 males, suggesting that genetic background and sex hormones influence FAM84A function . Methodological differences between studies, including variations in experimental systems (in vitro vs. in vivo), cell lines, and detection methods, can contribute to seemingly contradictory results. The genetic background of model systems, including the status of related pathways like Wnt/β-catenin, may significantly impact FAM84A's functional effects. To reconcile contradictory findings, researchers should perform parallel experiments using multiple model systems under identical conditions, employ complementary methods to validate findings, conduct comprehensive pathway analysis to identify context-dependent interacting factors, and consider dose-dependent effects. Thorough documentation of experimental conditions, transparent reporting of limitations, and consideration of negative results are essential for building a more complete understanding of FAM84A function across different biological contexts.
Studying FAM84A in patient samples requires careful consideration of collection, processing, and analysis methods. Fresh-frozen tissue samples are preferable for RNA and protein studies, though formalin-fixed paraffin-embedded (FFPE) samples can be used with appropriate extraction protocols. For RNA analysis, qRT-PCR remains the gold standard, with primers designed to specifically amplify FAM84A (such as forward 5'‐ATTCGGCTCGGGGTAGAG‐3' and reverse 5'‐TCTTCCTCATCATCCGAGAA‐3' for human FAM84A) . Expression should be normalized to stable reference genes like GAPDH . For protein detection, immunohistochemistry can visualize FAM84A expression patterns within tissue architecture, while tissue microarrays enable high-throughput analysis across multiple patient samples. Western blotting of tissue lysates provides quantitative assessment of protein levels . Patient cohort selection should consider factors such as cancer type, stage, treatment history, and outcome data to enable meaningful clinical correlations. Statistical analysis should employ appropriate methods based on data distribution, with non-parametric tests often being suitable for patient samples with inherent variability. Multivariate analysis should be conducted to account for confounding factors, and survival analysis can assess prognostic significance. Finally, validation in independent patient cohorts is essential for confirming findings.
Addressing the tissue-specific and context-dependent effects of FAM84A requires a comprehensive research strategy. First, researchers should systematically characterize FAM84A expression and function across multiple tissue types and cellular contexts. This includes analyzing both normal and pathological tissues, as well as considering factors like sex differences and genetic background. The marked differences in FAM84A induction between male and female mice, as well as between C3He and C57BL6 strains, highlight the importance of these factors . Comparative studies using identical methodologies across different tissue types or cancer models can help identify tissue-specific regulatory mechanisms and functional effects. When studying FAM84A in a specific tissue context, researchers should consider relevant physiological conditions, such as hormonal influences or metabolic states. For mechanistic studies, identifying tissue-specific binding partners and signaling networks is essential. This might involve tissue-specific proteomics approaches to identify differential interactomes. Additionally, conditional knockout or knockdown models that allow tissue-specific and temporal control of FAM84A expression can help dissect its function in particular contexts. Finally, integrating data across different experimental systems and developing computational models that account for context-specific variables will facilitate a more comprehensive understanding of FAM84A biology.
Developing FAM84A as a therapeutic target presents several significant challenges. Target specificity is a primary concern, as therapies must specifically modulate FAM84A without affecting related family members or proteins with similar domains. Since FAM84A appears to function as a scaffold or adaptor protein rather than an enzyme with a clearly defined active site, traditional small molecule inhibitor approaches may be challenging. Additionally, FAM84A's cytoplasmic localization means that therapeutic agents must penetrate the cell membrane to reach their target. The tissue-specific and context-dependent functions of FAM84A suggest that therapeutic strategies may need to be tailored to specific cancer types or molecular contexts, complicating development of broadly applicable approaches. For RNA-based therapeutics targeting FAM84A (such as siRNA or antisense oligonucleotides), effective delivery systems that can achieve sufficient tumor penetration while minimizing exposure to normal tissues will be crucial. Potential resistance mechanisms, including compensatory upregulation of related pathways, must be anticipated and addressed. Development of companion diagnostics to identify patients likely to benefit from FAM84A-targeted therapies will be important for clinical translation. Despite these challenges, the apparent involvement of FAM84A in multiple cancer types and its demonstrated roles in promoting proliferation, survival, and metastasis suggest it remains a promising target worth pursuing.
Several promising avenues for translational research on FAM84A merit exploration. Developing FAM84A as a diagnostic or prognostic biomarker represents an immediate opportunity, given the observed correlations between FAM84A expression and clinical parameters such as tumor size, lymph node metastasis, and advanced TNM stage in papillary thyroid carcinoma . This would require standardized detection methods and validation in large, diverse patient cohorts. RNA interference approaches targeting FAM84A have shown efficacy in reducing cancer cell proliferation and inducing apoptosis in experimental models , suggesting therapeutic potential. Development of delivery systems for siRNA or shRNA targeting FAM84A could advance this approach toward clinical applications. Since miR-874-3p negatively regulates FAM84A , miRNA-based therapeutics present another promising strategy. Identifying small molecule compounds that can modulate FAM84A expression or function through high-throughput screening could yield novel therapeutic candidates. Additionally, targeting the interaction between FAM84A and its binding partners could disrupt its oncogenic functions. Combination approaches, where FAM84A-targeted therapies are used alongside conventional treatments, warrant investigation for potential synergistic effects. Finally, exploring FAM84A's role in treatment resistance could reveal opportunities to enhance response to existing therapies.
Emerging technologies offer new opportunities to deepen our understanding of FAM84A biology. CRISPR-Cas9 genome editing enables precise manipulation of FAM84A, including complete knockout, introduction of specific mutations (like the serine 38 residue ), or creation of tagged versions for localization and interaction studies. Single-cell RNA sequencing could reveal cell-type specific expression patterns and heterogeneity within tumors. Spatial transcriptomics would provide insights into FAM84A expression in the context of tissue architecture and the tumor microenvironment. Advanced proteomics approaches, including proximity labeling techniques like BioID or APEX, could identify context-specific FAM84A interactomes. Cryo-electron microscopy might elucidate FAM84A's three-dimensional structure and reveal how it interacts with binding partners. Live-cell imaging with fluorescently tagged FAM84A could track its dynamics and localization in real-time. Organoid models derived from normal and cancerous tissues would provide more physiologically relevant systems for studying FAM84A function than traditional 2D cell cultures. Patient-derived xenografts that better preserve tumor heterogeneity and microenvironment could advance in vivo studies. Finally, computational approaches integrating multi-omics data could generate new hypotheses about FAM84A function and regulation in different cellular contexts.
While FAM84A research has primarily focused on cancer, its potential roles in non-cancer diseases warrant investigation. Given its reported effects on cell migration and morphology , FAM84A might influence processes requiring cellular migration and tissue remodeling, such as wound healing, tissue regeneration, or fibrotic diseases. The observed regulation of FAM84A by the nuclear xenobiotic receptor CAR suggests potential involvement in drug metabolism or toxicological responses, which could be relevant to liver diseases or drug-induced toxicities. FAM84A's apparent involvement in the Wnt/β-catenin signaling pathway points to possible roles in developmental disorders or degenerative conditions where this pathway is implicated. Its interaction with epithelial-mesenchymal transition processes suggests potential contributions to fibrotic disorders or inflammatory conditions involving tissue remodeling. Additionally, given the sex-dependent regulation observed in animal models , FAM84A might have sex-specific functions in normal physiology or disease processes. Systematic characterization of FAM84A expression across different disease states using tissue banks and public databases could reveal previously unrecognized associations. Functional studies in relevant model systems would then be required to determine whether these associations reflect causal relationships or secondary effects.
While direct evidence of FAM84A's interactions with the tumor microenvironment is limited in current literature, several plausible mechanisms warrant investigation. FAM84A's role in promoting epithelial-mesenchymal transition (EMT) suggests it may influence tumor-stroma interactions, as EMT is associated with altered secretion of cytokines, chemokines, and extracellular matrix components that reshape the microenvironment. The activation of Wnt/β-catenin signaling by FAM84A could affect paracrine signaling, as this pathway regulates expression of factors that modulate immune cell function and angiogenesis. Given that FAM84A promotes cancer cell proliferation and survival , it may indirectly influence the tumor microenvironment by increasing tumor burden and hypoxia, which trigger adaptive responses in stromal and immune cells. Co-culture experiments combining FAM84A-manipulated cancer cells with stromal components (fibroblasts, endothelial cells, immune cells) could reveal its effects on cellular crosstalk. Analysis of secretome changes upon FAM84A modulation might identify soluble factors mediating its influence on the microenvironment. In vivo models with intact immune systems would be valuable for studying FAM84A's effects on tumor immunity. Single-cell and spatial transcriptomics approaches could map FAM84A expression and associated pathway activities across different cellular populations within the tumor ecosystem.
Understanding the evolutionary conservation and divergence of FAM84A across species could provide insights into its fundamental biological functions. Comparative genomic analysis would reveal the evolutionary history of FAM84A, identifying when it emerged and how it has changed across lineages. Sequence conservation analysis could highlight functionally important domains that have been preserved through evolutionary pressure. For instance, the significance of the serine 38 residue identified in mouse FAM84A could be assessed by determining whether this site is conserved in other species. Examining FAM84A expression patterns across model organisms could reveal conserved and divergent aspects of its regulation and tissue distribution. The sex-dependent and strain-dependent expression patterns observed in mice raise interesting questions about whether similar regulatory patterns exist in other species, including humans. Functional studies in evolutionarily diverse model organisms (such as zebrafish, Drosophila, or C. elegans, if homologs exist) could determine whether FAM84A's roles in cell migration, proliferation, and cancer-related processes are evolutionarily conserved functions or more recent adaptations. This evolutionary perspective might also help identify functionally important interaction partners that have co-evolved with FAM84A across species.
FAM84A was identified through genome-wide cDNA microarray analysis, which revealed its frequent upregulation in colorectal cancer tissues . This gene was not expressed in any of the 23 normal tissues examined, except for the testis . The gene was cloned by researchers who noted its 44% amino acid identity with FAM84B and 34.5% identity with the C. elegans cuticle collagen-12 precursor .
The expression of FAM84A mRNA and protein has been confirmed to be upregulated in colorectal carcinomas compared to normal colonic mucosa . Additionally, FAM84A expression has been observed in bladder, pancreatic, and lung cancers . Immunocytochemical studies have localized FAM84A to the subplasma membrane region, particularly in areas where cells do not have contact with neighboring cells .
FAM84A plays a crucial role in cell migration and morphology. Studies have shown that exogenous expression of FAM84A increases cell motility in NIH3T3 cells . The phosphorylation of serine 38 of FAM84A is associated with changes in cell morphology, suggesting that this post-translational modification is important for its function . Wound healing and transwell assays have demonstrated that FAM84A significantly enhances cell migration and wound filling .
The upregulation of FAM84A in colorectal cancer and its role in enhancing cell motility suggest that it may play a critical role in cancer progression . Understanding the molecular mechanisms by which FAM84A contributes to cancer cell migration and invasion could provide insights into potential therapeutic targets for colorectal and other cancers.