CMC4 is a 93-amino acid recombinant protein (68 native residues + 25 His-tag) with a molecular mass of 10.4 kDa . Its sequence includes a conserved C-X(9)-C motif, characteristic of proteins involved in redox regulation and metal ion binding .
Property | Detail |
---|---|
Gene Location | Xq28 (Chromosome X) |
Transcript | NM_001018024.2 |
Protein Accession | NP_001018024.1 (UniProt: P56277) |
Expression | Highest in fetal testis; detectable in mitochondria |
CMC4 is implicated in:
Mitochondrial Regulation: Localizes to mitochondria, influencing apoptosis and inflammatory responses .
Cell Proliferation: Modulates pathways critical for Sertoli cell (SC) function and gonadotropin-releasing hormone regulation .
T-Cell Proliferation: Linked to t(X;14) translocations in mature T-cell proliferative disorders .
Deletions in CMC4 and adjacent FUNDC2 disrupt apoptosis and follicle-stimulating hormone (FSH) regulation, leading to HH. RNA-seq analyses show upregulated genes in SC regulation and inflammation in affected individuals .
CMC4 suppression via miR-126 inhibits tumor growth and migration, suggesting a role in cancer cell dynamics .
Disease | Mechanism | Source |
---|---|---|
Moyamoya Disease 1 | Vascular pathology linked to Xq28 defects | |
Rhabdomyolysis-Myalgia Syndrome | Mitochondrial dysfunction |
Production: Expressed in E. coli with >90% purity (SDS-PAGE) .
Storage: Stable at -20°C with 20% glycerol; requires carrier proteins (e.g., HSA/BSA) for long-term storage .
Product | Applications | Vendor |
---|---|---|
Anti-CMC4 Antibody (PRO-1769) | WB, IHC, ICC | Thermo Fisher |
Rabbit Polyclonal Anti-CMC4 | IHC, ICC-IF, WB | Atlas Antibodies |
The LOVD database documents 5 public variants in CMC4, including 4 unique DNA changes associated with clinical phenotypes . Skewed X-chromosome inactivation in female carriers mitigates phenotypic severity .
Current gaps include mechanistic insights into CMC4’s role in mitochondrial apoptosis and validation of its interactors. Functional studies using gene-editing models are needed to clarify its contributions to HH and cancer .
CMC4 is a gene located on chromosome Xq28, sharing a complex gene structure with MTCP1, including a common promoter and 5' exon. This gene occupies a critical region associated with various phenotypes when deleted, particularly hypogonadotropic hypogonadism (HH). Its location on the X chromosome makes it especially significant for understanding X-linked disorders and sex-specific developmental traits .
CMC4 protein levels are highest in fetal testis compared to other examined tissues, including adult testis. This suggests a developmentally regulated role, particularly in gonadal development. Interestingly, standard RNA-seq approaches may struggle to detect CMC4 expression due to annotation challenges, as evidenced by "0 raw RNA-seq count for CMC4 (Genecode gene ENSG00000182712) after mapping using STAR," despite substantial RNA-seq reads aligning to multiple CMC4 exons in control samples .
Tissue Type | Expression Level | Detection Method |
---|---|---|
Fetal Testis | High | Protein analysis |
Adult Testis | Moderate | Protein analysis |
Peripheral Blood | Low/Undetectable | RNA-seq |
Other Adult Tissues | Low/Undetectable | GTEx database |
CMC4 and MTCP1 share genomic elements, including a common promoter and 5' exon, but appear to have distinct functions. Research indicates that "MTCP1 is not expressed in normal tissues," suggesting CMC4 may be the functionally relevant gene in normal physiology. Literature inconsistencies exist, with one study referring to Cmc4 as "misnamed as Mtcp1" because the antibody used targeted peptides encoded by Cmc4, highlighting the importance of clear distinction between these genetic elements in research design .
Detection of CMC4 expression requires specialized approaches due to its complex gene structure and annotation challenges. Effective methodologies include:
Custom RNA-seq analysis pipelines that specifically target CMC4 exons
RT-qPCR with primers designed to unique regions, avoiding shared exons with MTCP1
Western blot analysis using validated antibodies specific to CMC4 protein
Tissue-specific in situ hybridization for spatial expression patterns
When analyzing expression data, researchers should be aware that "no read was observed on any exon including the undeleted exon 1 in the patient [with CMC4 deletion], probably due to degradation of the premature transcript" .
Initial functional studies suggest CMC4 may influence cell proliferation and mobility. A study using a nude mouse tumor model demonstrated that suppression of Cmc4 by miR-126 is involved in repression of tumor growth and migration. Given that "CMC4 protein level is highest in fetal testis among the examined tissues," researchers speculate it may play a critical role in testicular development and function .
Deletion of CMC4 on chromosome Xq28 has been associated with a constellation of clinical features, most notably hypogonadotropic hypogonadism (HH), short stature, and bilateral cataracts. Analysis of patient samples revealed that "up-regulated genes in the patient are enriched in SC regulation, gonadotropin-releasing pathway, apoptosis, and inflammatory response," consistent with extremely high FSH levels observed in affected individuals. The critical minimal region of overlap between patients has been narrowed to exonic deletions of CMC4 and MTCP1 .
When investigating CMC4's role in apoptosis, inflammation, and gonadotropin regulation, researchers should consider:
Multi-omic approaches to capture the full spectrum of pathway dysregulation
Time-course experiments to track dynamic changes following CMC4 perturbation
Cell type-specific analyses, particularly in gonadal tissues where CMC4 is highly expressed
Careful selection of experimental models that recapitulate human developmental contexts
Validation across multiple methodological platforms to overcome detection challenges
Research suggests that "loss of function of CMC4 results in dysregulation of apoptosis, inflammation and FSH," requiring comprehensive experimental designs that can address these interconnected pathways .
Based on current research, the following experimental models show promise for CMC4 functional studies:
Patient-derived samples with natural CMC4 deletions or mutations
CRISPR-Cas9 engineered cell lines with specific CMC4 modifications
Nude mouse models, previously used to study Cmc4 regulation by miR-126
In vitro gonadal differentiation models to investigate developmental roles
Induced pluripotent stem cells (iPSCs) differentiated toward relevant lineages
When designing DOE (Design of Experiments) approaches for these models, researchers should follow systematic methodology similar to those used in CMC drug development, where multiple variables are simultaneously evaluated to identify optimal conditions and key parameters .
Distinguishing direct from indirect effects of CMC4 deletion requires sophisticated experimental designs:
Temporal analysis of transcriptional and proteomic changes following acute CMC4 depletion
Rescue experiments reintroducing wild-type or mutant CMC4 variants
ChIP-seq or similar approaches to identify direct genomic targets if CMC4 functions as a transcriptional regulator
Protein-protein interaction studies to map the CMC4 interactome
Pathway inhibition experiments to delineate the hierarchy of dysregulated processes
Such approaches can help determine whether observed effects like "enrichment in SC regulation, gonadotropin-releasing pathway, apoptosis, and inflammatory response" represent direct or downstream consequences of CMC4 loss .
Several notable contradictions and knowledge gaps exist in CMC4 research:
Annotation discrepancies: Despite zero counts in GTEx databases, substantial RNA-seq reads align to CMC4 exons in control samples
Nomenclature confusion: Some studies have mislabeled CMC4 as MTCP1, complicating literature reviews
Functional inconsistencies: While some evidence suggests roles in cell proliferation, other data points to developmental pathways
Expression patterns: Questions remain about whether the high fetal testis expression represents the primary site of action
Researchers should be aware that "it is not clear how loss of function of CMC4 causes [HH]," highlighting the need for further mechanistic studies .
Design of Experiments (DOE) offers powerful advantages for CMC4 research by systematically exploring multiple experimental variables simultaneously:
Identify critical parameters potentially affecting CMC4 expression or function
Design factorial or response surface experiments to efficiently test parameter combinations
Establish clear response variables related to hypothesized CMC4 functions
Apply statistical analysis to identify significant factors and interactions
Use the results to optimize experimental conditions and define the operational space
As noted in pharmaceutical research, "DOE explores the simultaneous effect of multiple factors, offering a more holistic and effective process" compared to changing one factor at a time .
Given the challenges in detecting CMC4 expression, researchers should consider:
Custom bioinformatic pipelines for RNA-seq data that specifically target CMC4 exons
Validated antibodies and immunological techniques specific to CMC4 protein
Multiple reference genes for normalization in qPCR experiments
Spatial transcriptomics to map expression patterns across tissues
Single-cell approaches to identify cell-specific expression patterns
These approaches help overcome the annotation challenges noted in studies where "0 raw RNA-seq count for CMC4... is consistent with the complete lack of CMC4 gene expression track from the GTEx database of any tissue" despite evidence of expression through other methods .
Establishing causality between CMC4 deletion and phenotypes requires rigorous experimental approaches:
Generate precisely defined CMC4 mutations/deletions using CRISPR-Cas9
Perform rescue experiments with wild-type CMC4 in deletion models
Create allelic series with varying degrees of CMC4 function
Employ tissue-specific and inducible systems to control timing and location of CMC4 disruption
Validate findings across multiple model systems and human patient samples
This multi-faceted approach addresses the challenge that "it is not clear how loss of function of CMC4 causes [observed phenotypes]" and helps establish direct causality .
When analyzing CMC4 gene interactions, researchers should:
Distinguish between physical interactions (protein-protein) and genetic interactions
Control for shared regulatory elements between CMC4 and MTCP1
Consider developmental timing when interpreting interaction data
Validate interactions using multiple methodological approaches
Assess the functional consequences of disrupting specific interactions
These practices help address the complexity noted in studies where "CMC4 is associated with genes involved in SC regulation, gonadotropin-releasing pathway, apoptosis, and inflammatory response" .
A comprehensive risk assessment framework for CMC4 research includes:
Identification of critical quality attributes (CQAs) for experimental outcomes
Systematic evaluation of experimental parameters affecting those CQAs
Development of risk profiles for different experimental approaches
Implementation of control strategies focused on highest-impact variables
Continuous monitoring and refinement of methodologies
This approach mirrors strategies used in pharmaceutical development where "DOE provides a structured framework for risk assessment" by evaluating various parameters on critical quality attributes .
When facing contradictory CMC4 expression data, researchers should:
Compare methodological approaches used across studies (RNA-seq vs. protein detection)
Evaluate annotation versions and mapping algorithms used for sequence data
Consider tissue-specific or developmental timing differences
Assess the specificity of reagents used (primers, antibodies)
Perform targeted validation studies using multiple techniques
These steps address challenges exemplified by findings where "substantial RNA-seq reads were aligned to multiple exons of CMC4 in RNA-seq of mother and the controls" despite absence in standard databases .
Analysis of complex phenotypes associated with CMC4 deletion requires:
Multivariate statistical methods to capture interrelated phenotypic traits
Longitudinal analysis for developmental phenotypes
Careful selection of appropriate control groups
Power analysis to ensure sufficient sample sizes
Machine learning approaches for pattern recognition in complex datasets
These methods help characterize the multifaceted phenotypes where "up-regulated genes in the patient are enriched in SC regulation, gonadotropin-releasing pathway, apoptosis, and inflammatory response" .
Effective integration of multi-omic data requires:
Harmonized sample preparation and data collection protocols
Computational frameworks that account for different data types and scales
Network analysis approaches to identify functional connections
Validation of key findings across multiple data types
Pathway enrichment methods that incorporate diverse data sources
This integration helps elucidate the complex regulatory networks suggested by studies where loss of CMC4 affects multiple cellular pathways .
Thorough documentation of CMC4 research should include:
Detailed characterization of antibodies, including epitope information and validation data
Complete primer sequences and PCR conditions for expression studies
Comprehensive description of cell lines, including passage number and authentication
Clear identification of genetic constructs, including sequence verification
Precise recording of experimental conditions, particularly for developmental studies
This thoroughness addresses issues like those where "Cmc4 [was] misnamed as Mtcp1 in [a] study, since the antibody used is for the peptide encoded by Cmc4" .
Cross-platform collaborative studies benefit from:
Standardized protocols for sample preparation and data collection
Reference samples processed across all platforms for normalization
Data sharing agreements established before study initiation
Common data analysis pipelines or cross-validation of platform-specific approaches
Regular communication to address methodological challenges
These practices ensure robust findings across diverse experimental settings, particularly important given the complex nature of CMC4 function and detection .
Future studies investigating CMC4's developmental functions should consider:
Time-course analyses of CMC4 expression across developmental stages
Single-cell transcriptomics of developing tissues, particularly gonads
Lineage tracing in developmental models with CMC4 modifications
Interaction studies with known developmental regulators
Cross-species comparative analyses to identify conserved functions
These approaches address the observation that "CMC4 protein level is highest in fetal testis among the examined tissues" and may help explain its potential role in development .
Next-generation genetic engineering approaches for CMC4 research include:
Base editing for precise nucleotide modifications without double-strand breaks
Prime editing for flexible gene editing with minimal off-target effects
Inducible degron systems for temporal control of CMC4 protein levels
CRISPRi/CRISPRa for reversible modulation of CMC4 expression
CRISPR screening to identify genetic interactors of CMC4
These techniques provide unprecedented control and specificity for investigating CMC4 function beyond traditional knockout approaches .
Translational research directions for CMC4 include:
Development of biomarkers for conditions associated with CMC4 dysfunction
Screening of patient populations with unexplained HH for CMC4 mutations
Investigation of therapeutic approaches targeting downstream pathways affected by CMC4 loss
Development of model systems for drug screening
Exploration of gene therapy approaches for CMC4-related conditions
These translational efforts build on findings that link CMC4 deletion to specific clinical phenotypes including hypogonadotropic hypogonadism .
Advanced computational methods for CMC4 research include:
Protein structure prediction to understand CMC4 function
Systems biology modeling of pathways affected by CMC4
Machine learning analysis of multi-omic data to identify patterns
Virtual screening for compounds that might modulate CMC4-related pathways
Network analysis to place CMC4 in broader cellular contexts
These computational approaches complement experimental work, particularly useful given the challenges in directly studying CMC4 .
Researchers investigating CMC4 in human genetics should consider:
Appropriate informed consent for patient samples, especially for studies involving reproductive development
Privacy protections for genetic data from individuals with CMC4 variants
Thoughtful communication of findings to affected individuals and families
Equitable research practices that include diverse populations
Careful consideration of potential clinical applications and their implications
These ethical considerations ensure responsible advancement of knowledge about CMC4 and its role in human health and development .
The CMC4 gene is part of a bicistronic locus that also includes the MTCP1 gene. Both genes share a common promoter and a 5’ untranslated region (UTR), but they have distinct open reading frames (ORFs) and encode different proteins . The CMC4 gene consists of at least seven exons, with a complex gene structure that includes a long GC-rich sequence in the 5’ UTR .
The CMC4 protein, also known as p8 MTCP1, is a mitochondrial protein that plays a role in mature T-cell proliferation . It is involved in various cellular processes, including peroxisomal lipid metabolism . The protein is 68 amino acids long and is expressed in a cytoplasmic granular pattern in transfected cells .
Mutations or deletions in the CMC4 gene have been associated with several diseases, including Microcephalic Osteodysplastic Primordial Dwarfism, Type I, and Pitt-Hopkins Syndrome . Additionally, the gene has been implicated in certain T-cell lymphoproliferative diseases, where it is often involved in chromosomal translocations .
The study of CMC4 is crucial for understanding its role in T-cell proliferation and its potential implications in various diseases. Recombinant forms of the protein are used in research to study its function and interactions within the cell. Understanding the molecular mechanisms of CMC4 can provide insights into potential therapeutic targets for related diseases.