TMEM14A antibodies target the 99-amino acid transmembrane protein (10.7 kDa) encoded by the TMEM14A gene (synonyms: C6orf73, PTD011) . These antibodies are validated for applications including Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and ELISA .
Structure: Three transmembrane domains, confirmed via nuclear magnetic resonance spectroscopy .
Expression: Elevated in ovarian cancer tissues compared to normal tissues , and expressed in podocytes, endothelial cells, and distal tubular cells in kidneys .
Function: Inhibits apoptosis by suppressing mitochondrial outer membrane permeabilization and modulates energy metabolism (glycolysis, oxygen respiration) .
In Ovarian Cancer:
Protects glomerular filtration barrier integrity by maintaining podocyte viability .
Reduced expression precedes proteinuria onset in rat models .
Apoptosis Studies: Used to investigate mitochondrial membrane potential stabilization and Bax suppression .
Cancer Biomarker Research: Detects overexpression in ovarian, colon, and hepatocellular carcinomas .
Renal Pathology: Identifies TMEM14A depletion in proteinuria models .
Cell Cycle Regulation: Modulates G1/S transition via Cyclin D1, Cyclin E, and PCNA .
Metastasis: Upregulates MMP-2 and MMP-9 to enhance invasiveness .
Energy Metabolism: Drives glycolysis and oxygen respiration in ovarian cancer cells .
TMEM14A (Transmembrane protein 14A) is a 99 amino acid integral membrane protein with three transmembrane domains. Its structure has been identified through nuclear magnetic resonance spectroscopy. TMEM14A primarily localizes in mitochondria and belongs to the transmembrane (TMEM) protein family, several members of which have been identified as having oncogenic properties . Understanding its structure is essential for interpreting functional studies, as the three transmembrane domains suggest potential interaction with membrane-associated signaling pathways.
TMEM14A shows differential expression across tissues, with particularly high expression in podocytes within the kidney. Research demonstrates that TMEM14A mRNA expression is highest in differentiated podocytes compared to other cell types such as human embryonic kidney (HEK) cells and human umbilical vein endothelial cells (HUVEC) . Within the kidney, expression is higher in isolated glomeruli than in whole kidney tissue, suggesting enrichment in glomerular structures. Immunohistochemistry has also revealed TMEM14A expression in distal tubular cells. In pathological contexts, TMEM14A shows elevated expression in ovarian cancer tissues compared to para-carcinoma tissues .
At the cellular level, TMEM14A performs multiple functions. It has been implicated in preventing apoptosis by preserving mitochondrial membrane potential through Bax suppression . In ovarian cancer cells, TMEM14A has been shown to inhibit cell apoptosis while accelerating energy metabolism, including both glycolysis and oxygen respiration . These findings suggest that TMEM14A may serve as a metabolic regulator at the mitochondrial level. Additionally, TMEM14A has been positively correlated with c-MYC expression in ovarian cancer, with overexpression of c-Myc rescuing the function of TMEM14A in experimental models .
When selecting a TMEM14A antibody for research applications, several critical factors should be considered:
Application compatibility: Ensure the antibody has been validated for your specific application (WB, IHC, IF, ELISA). For example, commercially available polyclonal antibodies have been validated for multiple applications including ELISA, Western blot, immunohistochemistry, and immunofluorescence .
Species reactivity: Verify that the antibody recognizes TMEM14A from your species of interest. Some antibodies are specific to human TMEM14A, while others may have cross-reactivity with multiple species .
Epitope recognition: Consider which region of TMEM14A the antibody targets. For example, antibodies raised against amino acids 45-78 of human TMEM14A target a specific epitope that might be more accessible in certain applications .
Antibody format:
Host species (e.g., rabbit, goat) should be selected to avoid cross-reactivity with other antibodies in multi-color staining
Clonality (monoclonal vs. polyclonal) affects specificity and sensitivity
Conjugation status (unconjugated vs. fluorophore/enzyme-conjugated)
Validation evidence: Prioritize antibodies with published validation data including positive/negative controls and citation in peer-reviewed publications.
Based on published research, the following protocol has been successfully used for TMEM14A immunohistochemistry:
For Formalin-Fixed Paraffin-Embedded Tissue:
Section tissues at 4 μm thickness
Deparaffinize and dehydrate sections
Perform antigen retrieval by boiling in Tris/EDTA buffer for 10 minutes
Wash in PBS
Incubate with primary antibody diluted in 1% BSA in PBS:
Polyclonal goat anti-TMEM14A (such as Santa Cruz Biotech, sc-248899)
Dilution: 1:200 for rat tissue, 1:150 for human tissue
Incubate at 4°C overnight
Wash in PBS
Incubate for 30 minutes with secondary antibody:
Polyclonal Rabbit Anti-Goat Immunoglobulins/horseradish peroxidase
Wash in PBS
Detect immunoreactivity with diaminobenzidine
Counterstain with hematoxylin
For Cultured Cells:
Transfer confluent cells to small glasses in a 24-well plate
After 24 hours incubation, wash cells in PBS
Fixate using -20°C methanol
Wash in PBS
Block with 5% Normal Rabbit Serum (NRS) for 1 hour
Aspirate NRS and incubate with primary antibody diluted in 1% BSA in PBS at 4°C overnight
Continue with secondary antibody and detection as described above
Note that while this protocol works well for human and rat material, the quality of staining has been reported as insufficient for reliable assessment in zebrafish material .
Validating antibody specificity is crucial for reliable TMEM14A research. Implement these methodological approaches:
Positive and negative controls:
Use tissues or cell lines with known high expression (e.g., differentiated podocytes) as positive controls
Include tissues from TMEM14A knockout models or cells with TMEM14A knockdown as negative controls
Peptide competition assay:
Pre-incubate the antibody with excess immunizing peptide before application
Loss of signal confirms specificity for the target epitope
Cross-validation with multiple antibodies:
Compare staining patterns using antibodies targeting different TMEM14A epitopes
Consistent patterns increase confidence in specificity
Multiple detection methods:
Compare results between immunohistochemistry, Western blot, and immunofluorescence
Verify that molecular weight in Western blot corresponds to predicted TMEM14A size
Genetic manipulation validation:
Compare staining in wild-type versus TMEM14A-knockdown cells
Significant reduction in signal should be observed in knockdown samples
Species cross-reactivity testing:
TMEM14A has been identified as a potential oncogenic factor in ovarian cancer progression through multiple mechanisms. Research has demonstrated that TMEM14A is highly expressed in ovarian cancer tumors compared to para-carcinoma tissues, and this elevated expression correlates with higher mortality rates in patients .
Mechanistically, TMEM14A promotes ovarian cancer development by:
Inhibiting apoptosis: TMEM14A reduces cancer cell death, promoting tumor cell survival
Enhancing energy metabolism: TMEM14A accelerates both glycolysis and oxygen respiration, providing metabolic advantages to cancer cells
Interacting with oncogenic pathways: TMEM14A positively correlates with c-MYC expression, with overexpression of c-Myc rescuing the function of TMEM14A in experimental models
Functional studies using RNA interference and lentiviral-mediated vector systems have demonstrated that knockdown of TMEM14A suppresses the growth of human ovarian cancer cells by blocking glycolysis activity . These findings suggest TMEM14A may serve as both a diagnostic and prognostic biomarker for early detection of ovarian cancer and potentially as a therapeutic target.
Several robust methodological approaches have proven effective for studying TMEM14A in cancer cell lines:
Expression Modulation:
RNA interference (siRNA) for transient knockdown
Lentiviral-mediated vector systems for stable knockdown or overexpression
CRISPR-Cas9 for gene editing
Functional Assays:
Flow cytometric analysis to examine cell apoptosis
Seahorse XF24 analyzer to determine oxygen consumption and extracellular acidification
Cell proliferation assays to assess growth effects
Molecular Interaction Studies:
Chromatin immunoprecipitation assay to determine the connection between TMEM14A and transcription factors like c-Myc
Co-immunoprecipitation to identify protein-protein interactions
In Vivo Validation:
Xenograft mice models using transfected cancer cell lines
Immunohistochemical staining to determine expression patterns of TMEM14A and related factors in tumor tissues
For example, one study used CAOV3 ovarian cancer cells with lentivirus transfection, achieving over 80% transfection rate as verified by RT-qPCR after 72 hours . Cell culture conditions typically involve RPMI 1640 Medium with added antibiotics, insulin, transferrin, selenite, and 10% fetal bovine serum.
This prognostic value may stem from TMEM14A's dual role in inhibiting apoptosis and enhancing metabolic activity in cancer cells. By promoting cell survival and providing metabolic advantages, TMEM14A appears to contribute to more aggressive tumor behavior.
When analyzing TMEM14A expression for prognostic purposes, researchers should consider:
Expression levels relative to matched normal tissues
Correlation with established markers like c-Myc
Association with clinical variables including stage and grade
Standardization of quantification methods across studies
Importantly, TMEM14A has been observed to be deregulated in multiple cancer types, including hepatocellular carcinoma and colorectal cancer, suggesting its potential as a broader cancer biomarker beyond ovarian cancer .
TMEM14A has been identified as a critical protein required for maintaining the integrity of the glomerular filtration barrier (GFB). Research indicates that TMEM14A expression is diminished before the onset of proteinuria in spontaneously proteinuric rat models, suggesting a protective role in normal kidney function .
Experimentally, knocking down tmem14a mRNA translation in zebrafish embryos resulted in proteinuria without affecting tubular reabsorption, directly implicating TMEM14A in GFB maintenance . The molecular mechanisms through which TMEM14A protects the GFB may relate to its role in preventing apoptosis. Since podocyte apoptosis and detachment have been implicated in proteinuric renal diseases, TMEM14A's anti-apoptotic function may be critical for maintaining podocyte viability and thus GFB integrity.
Interestingly, TMEM14A expression patterns change with age and disease state. In healthy control rats (SHR), glomerular TMEM14A mRNA expression was significantly higher at younger ages (weeks 2, 4, and 6) compared to older ages (weeks 8 and 10), suggesting developmental regulation . In proteinuric models, this normal expression pattern is disrupted, with consistently lower expression at all time points.
Several approaches have been validated for silencing TMEM14A expression in different research models:
For Cell Culture Models:
RNA Interference (siRNA):
Culture cells to 80% confluence
Incubate in serum-free medium for 4 hours
Transfect with siRNA targeting TMEM14A using commercial transfection reagents
Confirm knockdown efficiency by RT-qPCR after 48-72 hours
Lentiviral-Mediated Knockdown:
Seed cells (e.g., 1.5 × 10^5 cells/well) in a 12-well plate and culture to 80% confluence
Incubate in serum-free medium for 4 hours
Transfect with lentiviruses carrying shRNA targeting TMEM14A
After 3 days, filter transfected cells using a 0.45 μM mesh
Concentrate the viral suspension at 70,000 × g at 4°C for 2 hours
Collect supernatant for viral titer determination
Culture target cells with diluted lentiviruses
Screen for transfection rate at 72 hours (aim for >80% transfection)
For Zebrafish Models:
Use morpholino injection to block mRNA translation of the zebrafish TMEM14A homologue (zgc:163080)
Verify knockdown through RT-qPCR
Assess functional effects using dextran tracer injection (3 and 70 kDa) to evaluate filtration barrier integrity
For quantifying knockdown efficiency by RT-qPCR, the following human TMEM14A primers have been validated:
Housekeeping genes such as GAPDH or HPRT1 should be used as internal controls for normalization.
Several experimental models have proven effective for studying TMEM14A function in kidney disease:
Rat Models:
The Dahl salt-sensitive rat has been used as a spontaneously proteinuric model where TMEM14A expression is diminished before proteinuria onset
Spontaneously hypertensive rats (SHR) serve as controls
These models allow temporal analysis of TMEM14A expression in relation to disease progression
Zebrafish Embryo Models:
Knockdown of the zebrafish TMEM14A homologue (zgc:163080) using morpholino injection
Functional assessment using dextran tracer injection (3 and 70 kDa) to evaluate filtration barrier integrity
Puromycin aminonucleoside (PAN) injection as a positive control for inducing proteinuria
Cell Culture Systems:
Immortalized podocytes for in vitro studies of TMEM14A function
Comparison with HEK293 and HUVEC cells to assess tissue-specific effects
Transfection experiments to modulate TMEM14A expression
Human Tissue Analysis:
Immunohistochemical staining of TMEM14A in kidney biopsies from patients with various proteinuric renal diseases
Comparative analysis of expression patterns in diseased versus healthy tissue
The table below summarizes genes found to be differentially expressed in proteinuric rat models, including TMEM14A:
| Gene name | Symbol | Region of rat chromosome | Fold change |
|---|---|---|---|
| Aldo-keto reductase family 1, member B8 | Akr1b8 | 4q22 | -4.5 |
| Similar to interferon regulatory factor 10 | RGD1562711 | 3q41 | -3.9 |
| Acyl-Coenzyme A oxidase 2, branched chain | Acox2 | 15p14 | -3.7 |
| Similar to RIKEN cDNA 4921520P21; DMRTC1 | LOC363483 | Xq31 | -3.4 |
| Transmembrane protein 14A | Tmem14a | 9q13 | -3.0 |
This table highlights that TMEM14A showed a 3-fold decrease in expression in proteinuric models .
Proper normalization of TMEM14A expression data is critical for accurate interpretation across different experimental conditions. Based on published methodologies, the following approaches are recommended:
For RT-qPCR Data:
Reference Gene Selection:
Calculation Method:
For Protein Expression Data:
Western Blot Normalization:
Normalize to housekeeping proteins such as β-actin, GAPDH, or α-tubulin
Consider total protein normalization methods for more accurate quantification
Immunohistochemistry Quantification:
Use digital image analysis with standardized acquisition settings
Quantify staining intensity and/or percentage of positive cells
Include internal controls within each experiment
The table below provides validated primer sequences for TMEM14A expression analysis:
| Name | Symbol | mRNA sequence | Forward primer | Reverse primer |
|---|---|---|---|---|
| Transmembrane protein 14A (human) | TMEM14A | NM_014051.3 | TTTGGTTATGCAGCCCTCGT | ATAGCCGGCCAAACATCCAA |
| Glyceraldehyde-3-phosphate dehydrogenase (human) | GAPDH | NM_002046 | TGGTCACCAGGGCTGCTT | AGCTTCCCGTTCTCAGCCTT |
| Hypoxanthine-guanine phosphoribosyltransferase 1 (human) | HPRT1 | NM_000194.2 | TGACACTGGCAAAACAATGCA | GGTCCTTTTCACCAGCAAGCT |
These validated primers ensure reliable quantification of TMEM14A expression .
When faced with contradictory TMEM14A expression data across studies or experimental conditions, researchers should implement systematic approaches to reconcile these differences:
1. Methodological Assessment:
Detection Method Differences: Compare RT-qPCR, Western blot, and immunohistochemistry results, as each method has different sensitivities and specificities
Antibody Specificity: Different antibodies may recognize different epitopes or isoforms of TMEM14A
Protocol Variations: Consider differences in tissue processing, fixation methods, and staining protocols
2. Biological Variables:
Cell Type Specificity: TMEM14A is differentially expressed across cell types, with highest expression in differentiated podocytes
Developmental Stages: TMEM14A expression changes with development, with higher expression at younger ages in rat models
Disease Context: TMEM14A shows elevated expression in ovarian cancer tissues but decreased expression in proteinuric kidney disease models
3. Analytical Approaches:
Meta-analysis: Pool data across studies with similar methodologies to identify consistent trends
Subgroup Analysis: Stratify results by relevant variables (tissue type, disease stage, age)
Correlation with Functional Outcomes: Relate expression data to functional readouts
4. Validation Strategies:
Orthogonal Methods: Confirm findings using multiple independent techniques
Alternative Models: Test expression in multiple model systems
Genetic Manipulation: Use knockdown/overexpression to establish causal relationships
Researchers analyzing TMEM14A expression across different tissues should be aware of several potential pitfalls:
1. Tissue Heterogeneity:
TMEM14A shows differential expression across cell types within the same organ. For example, in the kidney, it is primarily expressed in podocytes but also in distal tubular cells . Using whole-tissue homogenates without accounting for this heterogeneity can mask cell-specific changes in expression.
3. Disease-Induced Cellular Changes:
In disease states, the cellular composition of tissues can change (e.g., through inflammatory infiltrates or fibrosis). These changes can alter the apparent expression of TMEM14A independent of actual per-cell expression changes.
4. Antibody Specificity Issues:
Different antibodies may recognize different epitopes or isoforms of TMEM14A, potentially leading to discrepant results. The quality of staining has been reported as insufficient for reliable assessment in some species (e.g., zebrafish) , highlighting the importance of antibody validation.
5. Post-Transcriptional Regulation:
mRNA and protein levels may not correlate due to post-transcriptional regulation. Studies have shown that TMEM14A protein expression was lower in Dahl rats than in SHR at all time points after 2 weeks of age, which was significantly so at 4 and 8 weeks of age, suggesting complex regulation .
6. Experimental Variability:
Technical variables such as tissue processing, fixation methods, and staining protocols can significantly impact detected expression levels. For example, different dilutions of the same antibody have been recommended for different tissues (1:200 for rat tissue, 1:150 for human tissue) .
TMEM14A research is expanding into several promising directions that may significantly impact both basic science and clinical applications:
Biomarker Development:
Therapeutic Target Exploration:
Modulation of TMEM14A to inhibit cancer cell metabolism and growth
Preservation or enhancement of TMEM14A expression to protect podocyte function in kidney disease
Development of small molecule inhibitors or activators targeting TMEM14A-dependent pathways
Mechanistic Investigations:
Elucidation of the complete signaling network connecting TMEM14A to c-Myc and other oncogenic pathways
Clarification of TMEM14A's role in mitochondrial function and cellular energy metabolism
Investigation of potential post-translational modifications affecting TMEM14A function
Advanced Imaging Applications:
Development of live-cell imaging techniques to monitor TMEM14A dynamics in real-time
Super-resolution microscopy to investigate TMEM14A's precise subcellular localization
Correlative light and electron microscopy to link TMEM14A localization with ultrastructural features
These emerging applications underscore the importance of continuing to refine TMEM14A detection methodologies and expand our understanding of its biological functions across different tissues and disease states.
Several methodological advances would significantly enhance TMEM14A research:
Improved Antibody Development:
Generation of more specific monoclonal antibodies targeting different TMEM14A epitopes
Development of antibodies with broader species cross-reactivity, particularly for zebrafish models where current antibodies show limited efficacy
Creation of phospho-specific antibodies to detect potential regulatory post-translational modifications
Advanced Genetic Models:
CRISPR-engineered cell lines and animal models with conditional TMEM14A knockout/knockin
Reporter systems to monitor TMEM14A expression in real-time
Humanized animal models expressing human TMEM14A variants
Structural Biology Approaches:
High-resolution crystal or cryo-EM structures of TMEM14A in different functional states
Molecular dynamics simulations to understand conformational changes
Protein-protein interaction mapping through proximity labeling approaches
Single-Cell Technologies:
Single-cell RNA sequencing to map TMEM14A expression across all cell types in tissues of interest
Single-cell proteomics to correlate TMEM14A protein levels with mRNA expression
Spatial transcriptomics to maintain tissue context while assessing cell-specific expression
Functional Assays:
Development of high-throughput screening assays for TMEM14A modulators
More sensitive assays to quantify TMEM14A's effects on mitochondrial function
Standardized methods to assess TMEM14A's impact on the glomerular filtration barrier
These methodological advances would address current limitations in TMEM14A research and facilitate more comprehensive investigations into its functions across different biological contexts.