The DNAJC19 gene spans 6,065 base pairs on chromosome 3q26.33 and encodes a 6.29 kDa protein (59 amino acids) with a unique structure:
J-domain: Located at the C-terminal (unlike most DnaJ proteins), enabling interaction with mtHsp70 ATPase.
Transmembrane domain: Anchors the protein to the inner mitochondrial membrane.
Mitochondrial localization: Confers membrane-bound function, distinct from cytosolic DnaJ proteins .
Feature | Detail | Source |
---|---|---|
Gene symbol | DNAJC19 | |
Chromosome | 3q26.33 | |
mRNA length | 6065 bp | |
Protein weight | 6.29 kDa | |
Transmembrane domains | 1 (inner mitochondrial membrane) |
DNAJC19 (TIM14) is a core component of the TIM23 complex, facilitating ATP-dependent import of nuclear-encoded mitochondrial proteins into the matrix. It stimulates mtHsp70 ATPase activity to drive this process .
Interacts with prohibitins: Regulates cardiolipin synthesis, a phospholipid critical for mitochondrial fusion/fission and apoptosis.
Maintains mitochondrial morphology: Disruption leads to altered membrane structure and bioenergetic dysfunction .
Interaction Partner | Role | Source |
---|---|---|
TIMM44 | TIM23 complex component | |
mtHsp70 | ATP-dependent translocation | |
PHB2 | Cardiolipin remodeling |
DNAJC19 mutations cause autosomal recessive DCMA, characterized by:
Cardiac: Dilated cardiomyopathy, noncompaction, QT prolongation.
Neurological: Cerebellar ataxia, developmental delay.
Metabolic: 3-methylglutaconic aciduria type V (3-MGA5), microcytic anemia, growth failure .
DNAJC19 overexpression promotes non-small cell lung cancer (NSCLC) progression via AKT1 signaling:
Mechanism: Silencing DNAJC19 reduces AKT1 expression, inhibiting cell migration and metastasis.
Therapeutic potential: Targeting DNAJC19 may offer novel strategies for NSCLC treatment .
The LOVD database lists 9 variants associated with DCMA, including missense and frameshift mutations . Clinical testing involves sequencing and urinary organic acid analysis (e.g., 3-MGA5) .
DNAJC19 encodes DnaJ Heat Shock Protein Family (Hsp40) Member C19, an inner mitochondrial membrane protein that plays a crucial role in mitochondrial protein import machinery . The protein contains a conserved DnaJ interaction domain and is homologous to the yeast protein Pam18/Tim14 (yPam18) . Functionally, DNAJC19 regulates cardiolipin remodeling by interacting with protein complexes known as prohibitins that form protein scaffolds and lipids in the inner mitochondrial membrane, which is essential for mitochondrial morphogenesis and metabolism . The gene consists of three isoforms: isoform 1 represents the full-length transcript (525 nucleotides), isoform 2 lacks the transmembrane domain due to an alternative start codon, and isoform 3 lacks the DnaJ domain due to exon 4 deletion .
DNAJC19 expression regulation varies across tissues, with particularly high expression in cardiac tissue due to the heart's significant mitochondrial density (approximately 35% of cardiomyocyte volume) . While the search results don't provide comprehensive data on tissue-specific expression patterns, quantitative PCR analyses have shown that DNAJC19 mRNA can be detected in peripheral blood mononuclear cells (PBMCs) . Understanding tissue-specific regulation requires analyzing transcript levels across various tissues using methods such as RT-qPCR with tissue-specific normalization to housekeeping genes like GAPDH, as demonstrated in published research .
Several pathogenic variants of DNAJC19 have been identified in human populations, with the original splice site mutation (NM_145261.4):c.130-1G > C being one of the most well-characterized . This splice site mutation results in loss of the full-length transcript. Other identified pathogenic variants include:
A homozygous frameshift variant c.159del (Phe54Leufs*5) in exon 4, resulting in a premature stop codon four positions downstream
Biallelic mutations c.[131_140del];[137_138insAGTATAATTGCC] generated through CRISPR/Cas9, both leading to frameshift and premature stop codons
All these variants result in loss of the full-length DNAJC19 protein or critical functional domains, suggesting a loss-of-function mechanism in the associated disorders .
For comprehensive identification of DNAJC19 variants, a multi-tiered approach is recommended:
Whole Exome Sequencing (WES): Serves as an initial screening tool to identify potential variants in DNAJC19 and related genes .
Confirmatory Sanger Sequencing: Essential for validating variants identified through WES and establishing their precise position and nature .
RT-PCR Analysis: Critical for assessing the impact of splice site mutations on transcript structure and abundance .
Quantitative PCR (qPCR): Enables measurement of transcript levels to determine if nonsense-mediated decay is occurring in patient samples .
For example, in a study identifying a novel homozygous frameshift variant (c.159del), researchers first employed WES followed by Sanger sequencing confirmation, then used RT-qPCR to demonstrate substantially reduced DNAJC19 mRNA expression in the patient compared to controls .
Distinguishing pathogenic from benign DNAJC19 variants requires a multi-faceted approach:
Functional Assays: Generate iPSC-derived cardiomyocytes to test the impact of variants on mitochondrial morphology, respiration, and cardiac function .
Expression Analysis: Measure mRNA and protein expression levels in patient samples to determine if the variant affects expression or stability .
Domain-specific Analysis: Assess whether the variant affects conserved functional domains, particularly the DnaJ domain which is critical for protein function .
Co-segregation Studies: Evaluate whether the variant segregates with disease in affected families .
Population Frequency Data: Check variant frequency in population databases to determine rarity.
Research has shown that variants causing loss of the DnaJ domain consistently demonstrate pathogenicity through altered mitochondrial structure and function .
Creating accurate DNAJC19 mutation models presents several technical challenges:
Embryonic Lethality: Complete DNAJC19 knockout may cause embryonic lethality due to its critical role in mitochondrial function, necessitating inducible or tissue-specific approaches.
Isoform Complexity: DNAJC19 has three isoforms, making it challenging to target specific isoforms without affecting others .
Tissue-Specific Effects: DNAJC19 mutations may have different effects in different tissues, requiring multiple model systems.
Off-Target Effects: CRISPR/Cas9 modification, while effective for generating biallelic truncating variants as demonstrated in research, carries risks of off-target modifications .
Phenotypic Variability: The extreme heterogeneity observed in patients with DNAJC19 mutations makes it difficult to establish definitive genotype-phenotype correlations .
Researchers have successfully addressed some of these challenges by using gene-edited induced pluripotent stem cells (iPSCs) that can be differentiated into cardiomyocytes, providing a human-relevant model system .
Optimizing iPSC-derived cardiomyocytes (iPSC-CMs) for DNAJC19 research requires attention to several factors:
Maturation Protocols: Standard differentiation protocols may yield immature cardiomyocytes. Extended culture periods (60+ days) improve maturation and mitochondrial network development, as used in published research .
Isogenic Controls: Generate gene-edited control lines from the same genetic background to minimize confounding variables .
Functional Assays: Incorporate comprehensive assessments including:
Multi-lineage Differentiation: Compare effects in different cell types derived from the same iPSC lines to identify cardiac-specific effects.
Research has demonstrated that iPSC-CMs can successfully recapitulate DCMA-associated phenotypes, including mitochondrial fragmentation, abnormal cristae formation, and altered calcium handling .
Assessment of mitochondrial dysfunction in DNAJC19 mutant cells should employ a comprehensive battery of techniques:
Structural Analysis:
Bioenergetic Assessment:
ROS and Membrane Potential:
Metabolic Substrate Utilization:
Research has shown that DNAJC19 mutant cells exhibit increased OCRs, elevated ROS production, and altered substrate utilization, providing multiple parameters for comprehensive assessment .
For effective measurement of DNAJC19 protein localization and interactions, researchers should employ:
Subcellular Fractionation:
Isolate mitochondrial, cytosolic, and nuclear fractions to determine localization of DNAJC19 protein
Western blot analysis with fraction-specific markers to confirm purity
Co-immunoprecipitation (Co-IP):
Identify interaction partners, particularly with prohibitins and other components of the protein import machinery
Use crosslinking approaches for transient interactions
Immunofluorescence Microscopy:
Proximity Ligation Assays:
Detect and visualize protein-protein interactions in situ
Particularly useful for confirming interactions with proposed partners in the cardiolipin remodeling pathway
Published research has demonstrated mislocalization of DNAJC19 in mutant cells, with some signal appearing in nuclei rather than exclusively in mitochondria, highlighting the importance of these techniques .
DNAJC19 mutations lead to cardiac dysfunction through a cascade of interrelated mechanisms:
Mitochondrial Structural Abnormalities:
Bioenergetic Dysfunction:
Cellular Stress Responses:
Cardiac Functional Impairment:
These combined defects compromise cardiac energetics and contractile function, ultimately leading to dilated cardiomyopathy with potential arrhythmogenic complications .
The relationship between DNAJC19 dysfunction and 3-methylglutaconic aciduria (3-MGA) involves disruption of mitochondrial metabolic pathways:
Cardiolipin Remodeling: DNAJC19 regulates cardiolipin remodeling by interacting with prohibitins in the inner mitochondrial membrane . Disruption of this process affects mitochondrial membrane composition and function.
Metabolic Pathway Alterations: Dysfunctional mitochondria with abnormal cristae structure impact metabolic flux through the TCA cycle and associated pathways .
Leucine Metabolism: 3-methylglutaconic acid is an intermediate in leucine catabolism. Mitochondrial dysfunction may impair this pathway, leading to accumulation of 3-methylglutaconic acid.
Shared Pathophysiology with Barth Syndrome: DCMA shares similarities with X-linked Barth syndrome, which is also characterized by 3-MGA and is caused by mutations in the tafazzin (TAZ) gene involved in cardiolipin remodeling .
Diagnostic approaches should include measurement of urinary organic acids to detect elevated excretion of 3-methylglutaconic acid and 3-methylglutaric acid, which serve as biochemical markers of the disorder .
The neurological manifestations in patients with DNAJC19 mutations, particularly cerebellar ataxia, likely stem from:
Mitochondrial Dependency in Neurons: Neurons have high energy demands and are particularly dependent on mitochondrial function, making them vulnerable to DNAJC19 deficiency.
Cerebellum-Specific Vulnerability: The cerebellum contains high-frequency firing Purkinje cells with extensive dendritic arbors requiring efficient energy production and calcium handling, both of which are compromised by DNAJC19 mutations .
Altered Calcium Homeostasis: Mutant DNAJC19 cells show elevated diastolic Ca²⁺ concentrations and abnormal Ca²⁺ kinetics , which may particularly affect cerebellar neuronal circuits that rely on precise calcium signaling.
ROS-Mediated Neuronal Damage: Increased reactive oxygen species production observed in DNAJC19 mutant cells may cause cumulative oxidative damage to neurons, contributing to progressive ataxia.
Developmental Effects: DNAJC19's role in mitochondrial import and biogenesis may affect neuronal development and circuit formation during critical periods.
Research models specifically examining cerebellar neurons derived from DNAJC19 mutant iPSCs would be valuable for further elucidating these mechanisms.
Therapeutic approaches targeting mitochondrial function for DNAJC19 mutations could include:
Antioxidant Therapies:
Metabolic Modulation:
Address altered substrate utilization with agents promoting fatty acid oxidation
Consider ketogenic diets to provide alternative energy substrates
Monitor effectiveness through metabolic flux analysis and cardiac function tests
Mitochondrial Dynamics Regulation:
Target mitochondrial fragmentation with compounds that promote fusion
Evaluate effects on mitochondrial network integrity and cristae structure
Cardiolipin-Targeted Approaches:
Gene Therapy/Editing:
Research into these approaches would benefit from the iPSC-CM model systems already established, which demonstrate key disease phenotypes and could serve as platforms for therapeutic screening .
Understanding genotype-phenotype correlations in DNAJC19-related disorders faces several limitations:
Phenotypic Heterogeneity:
Limited Case Numbers:
Modifier Genes:
Unidentified genetic modifiers may influence the phenotypic expression of DNAJC19 mutations
Whole genome sequencing and systems biology approaches would be needed to identify these factors
Environmental Factors:
Non-genetic factors including diet, exercise, and environmental stressors may influence disease manifestation
These factors are difficult to control for in human studies
Tissue-Specific Effects:
Different tissues may have varying thresholds for mitochondrial dysfunction
Research using multi-tissue models derived from the same patient iPSCs could help address this limitation
Future research directions should include comprehensive phenotyping of larger cohorts, combined with whole genome sequencing and multi-tissue modeling to better understand these correlations .
DNAJC19 interactions with other mitochondrial protein import machinery components involve complex molecular networks:
Prohibitin (PHB) Complex Interaction:
DNAJC19 interacts with prohibitins that form protein scaffolds in the inner mitochondrial membrane
This interaction is crucial for cardiolipin remodeling and mitochondrial morphogenesis
Research approaches should include co-immunoprecipitation and proximity ligation assays to map interaction domains
TIM23 Complex Associations:
Based on homology to yeast Pam18/Tim14, DNAJC19 likely associates with the TIM23 translocase complex
Research should explore if DNAJC19 mutations affect protein import efficiency using in vitro import assays
OPA1 Processing:
J-protein Co-chaperone Function:
Investigation of these interactions requires a combination of proteomics approaches, including BioID or APEX proximity labeling to identify the complete interactome of DNAJC19 in mitochondria.
Methodological approaches to identify therapeutic targets for DNAJC19-related disorders should include:
High-throughput Screening Platforms:
Multi-omics Integration:
Combine transcriptomics, proteomics, and metabolomics data from patient samples and model systems
Identify dysregulated pathways and potential points for therapeutic intervention
Use systems biology approaches to model pathway interactions
CRISPR-based Genetic Screens:
Perform genome-wide or targeted CRISPR screens in DNAJC19 mutant cells
Identify genetic modifiers that rescue cellular phenotypes
Validate hits as potential therapeutic targets
Patient-derived Organoids:
Develop cardiac organoids from patient iPSCs for more complex tissue-level screening
Evaluate compounds in a three-dimensional context that better recapitulates tissue architecture
Comparative Studies with Related Disorders:
The iPSC-CM models already established provide an excellent foundation for implementing these approaches, as they demonstrate key disease phenotypes including mitochondrial dysfunction, altered calcium handling, and contractile abnormalities .
Parameter | DNAJC19 Mutant Cells | Control Cells | Significance | Method of Assessment |
---|---|---|---|---|
Oxygen Consumption Rate (OCR) | Increased | Baseline | Higher electron transport chain activity | Seahorse XF Analyzer |
Extracellular Acidification Rate (ECAR) | Increased | Baseline | Altered metabolic flux | Seahorse XF Analyzer |
Fatty Acid Uptake | Decreased | Baseline | Altered substrate utilization | Radioactive tracer studies |
Glucose Utilization | Increased | Baseline | Compensatory metabolism | Metabolic flux analysis |
Reactive Oxygen Species (ROS) | Increased | Baseline | Oxidative stress | Fluorescence-based assays |
Mitochondrial Membrane Potential | Elevated | Baseline | Altered mitochondrial function | Potential-sensitive dyes |
DnaJ (Hsp40) Homolog, Subfamily C, Member 19, also known as DNAJC19, is a protein encoded by the DNAJC19 gene in humans. This protein is a member of the DnaJ heat shock protein family (Hsp40), which plays a crucial role in the cellular response to stress by acting as molecular chaperones. These chaperones assist in the proper folding of proteins, prevention of protein aggregation, and the refolding of misfolded proteins.
The DNAJC19 gene is located on chromosome 3 and is a protein-coding gene. The protein encoded by this gene is involved in the ATP-dependent transport of transit peptide-containing proteins from the inner cell membrane to the mitochondrial matrix . The DNAJC19 protein is also known by several aliases, including TIM14, Mitochondrial Import Inner Membrane Translocase Subunit TIM14, and Pam18 .
DNAJC19 functions as a mitochondrial co-chaperone, forming a complex with prohibitins to regulate cardiolipin remodeling . Cardiolipin is a unique phospholipid that is essential for the optimal function of several mitochondrial enzymes. The DNAJC19 protein may also be a component of the PAM complex, which is required for the translocation of transit peptide-containing proteins from the inner membrane into the mitochondrial matrix in an ATP-dependent manner .
Mutations or defects in the DNAJC19 gene are associated with a rare metabolic disorder known as 3-methylglutaconic aciduria type 5 (MGA5), also referred to as dilated cardiomyopathy with ataxia (DCMA) . This disorder is characterized by a combination of symptoms, including dilated cardiomyopathy, ataxia, and elevated levels of 3-methylglutaconic acid in the urine. The condition is inherited in an autosomal recessive manner.
Recombinant DNAJC19 protein is used in various research applications to study its function and role in mitochondrial biology. Understanding the mechanisms by which DNAJC19 operates can provide insights into the development of therapeutic strategies for conditions associated with mitochondrial dysfunction.