ATP5D Human

ATP Synthase Subunit D, Mitochondrial Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of ATP5D

Gene and Protein Characteristics

  • Gene Location: Chromosome 19p13.3 (3,075 base pairs) .

  • Protein: 17.5 kDa, 168-amino-acid subunit of the F1 catalytic core in mitochondrial ATP synthase .

  • Domain Organization:

    • F1 Domain: Catalytic core with α3β3 hexamer and single γ, δ, ε subunits.

    • Central Stalk: Connects F1 to the proton channel (F0) for rotational coupling .

Key Pathways

  • Respiratory electron transport (GeneCards).

  • Mitochondrial biogenesis .

Clinical Implications of ATP5D Mutations

Genetic Disorders

  • Mitochondrial Complex V Deficiency:

    • Mutations: c.245C>T (p.Pro82Leu), c.317T>G (p.Val106Gly) disrupt F1F0 assembly, reducing ATPase activity .

    • Phenotypes: Episodic metabolic crises, lactic acidosis, hyperammonemia, and hypoglycemia .

Neurodegenerative and Metabolic Diseases

  • ALS Pathogenesis: Reduced ATP5D expression correlates with mitochondrial synaptic dysfunction .

  • Cancer Metabolism:

    • Glycolysis Regulation: m1A RNA methylation stabilizes ATP5D transcripts via YTHDF1, enhancing glycolysis in cervical cancer .

    • Prognostic Marker: Elevated ATP5D levels associate with poor survival in cervical cancer .

Table 1: Clinically Significant ATP5D Mutations

MutationClinical PhenotypeFunctional Impact
c.245C>TNeonatal metabolic decompensationImpaired F1F0 assembly
c.317T>GChildhood encephalopathyReduced complex V activity

Research Findings in Disease Contexts

Cancer Metabolism

  • m1A Methylation: Demethylase ALKBH3 knockdown reduces ATP5D translation, suppressing glycolysis (↓ lactate, ATP) .

  • Therapeutic Target: CRISPR-mediated ATP5D demethylation increases ATP production, suggesting metabolic reprogramming strategies .

Male Fertility

  • Knockout Models: ATP5D-deficient mice exhibit prolonged mating intervals, impaired spermatogenesis, and testicular atrophy .

  • Biomarker Potential: Seminal plasma ATP5D levels are significantly reduced in idiopathic male infertility .

Subcellular Localization and Expression

  • Mitochondrial Localization: Inner membrane-associated, confirmed by immunofluorescence .

  • Tissue Expression: Ubiquitous, with high levels in metabolically active tissues (liver, heart) .

Future Directions

  • Therapeutic Strategies: Targeting ATP5D in cancers with dysregulated glycolysis (e.g., cervical, hepatocellular).

  • Diagnostic Development: ATP5D as a biomarker for mitochondrial disorders or male infertility screening.

Product Specs

Introduction
F-ATPase delta (ATP5D), also known as ATP synthase subunit delta, plays a crucial role in ATP synthesis by utilizing a proton gradient across the inner mitochondrial membrane during oxidative phosphorylation. ATP synthase comprises two linked multi-subunit complexes: F1, the soluble catalytic core, and Fo, the membrane-bound component responsible for proton translocation. The F1 complex consists of five subunits (alpha, beta, gamma, delta, and epsilon) with a stoichiometry of 3:3:1:1:1. In contrast, the Fo complex primarily comprises subunits a, b, and c. ATP5D is an integral component of the catalytic core (F1).
Description
This product consists of a non-glycosylated polypeptide chain of ATP5D, produced in E. coli. It encompasses amino acids 23 to 168 of the human ATP5D protein and has a molecular weight of 17.3 kDa. A 21 amino acid His-tag is fused to the N-terminus to facilitate purification. The protein has been purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
The product is supplied as a 0.25 mg/ml solution of ATP5D in a buffer containing 20mM Tris-HCl (pH 8.0), 20% glycerol, and 0.1M NaCl.
Stability
While the recombinant human ATP5D protein remains stable for up to one week at 4°C, it is recommended to store it at -18°C or lower. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the ATP5D protein is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
ATP synthase subunit delta mitochondrial, F-ATPase delta subunit, ATP5D.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAEAAAAPAA ASGPNQMSFT FASPTQVFFN GANVRQVDVP TLTGAFGILA AHVPTLQVLR PGLVVVHAED GTTSKYFVSS GSIAVNADSS VQLLAEEAVT LDMLDLGAAK ANLEKAQAEL VGTADEATRA EIQIRIEANE ALVKALE.

Q&A

What is ATP5D and what is its role in cellular bioenergetics?

ATP5D (ATP5F1D) is a subunit of mitochondrial ATP synthase, specifically part of the F1 complex (δ subunit). It plays a crucial role in coupling proton translocation across the inner mitochondrial membrane to ATP production through oxidative phosphorylation. This subunit is essential for proper assembly and function of ATP synthase (Complex V), the final enzyme in the electron transport chain responsible for generating ATP . Research methodologies to study ATP5D function include blue native PAGE for complex assembly analysis, respirometry for measuring ATP production capacity, and structural biology approaches to determine its position within the ATP synthase complex.

What are the characterized pathogenic variants of ATP5D in humans?

Two significant pathogenic homozygous missense variants in ATP5F1D have been well-characterized: c.245C>T (p.Pro82Leu) and c.317T>G (p.Val106Gly) . These mutations affect conserved residues and result in impaired assembly of the F1FO ATP synthase complex. Researchers investigating ATP5D variations should employ whole exome sequencing or targeted gene panels with appropriate bioinformatic pipelines to identify novel variants. Functional validation of newly identified variants requires fibroblast studies, enzyme activity assays, and potentially model organism complementation assays.

How is ATP5D expression measured in experimental and clinical samples?

Multiple methodologies can be employed to measure ATP5D at transcript and protein levels:

  • ELISA: Effectively used to detect ATP5D protein in seminal plasma of fertile versus infertile males

  • qRT-PCR: For quantifying ATP5D mRNA expression in tissue samples

  • Western blotting: For protein detection and semi-quantitative analysis

  • Immunohistochemistry: For visualizing ATP5D distribution in tissue sections

  • Mass spectrometry: For precise quantification and detection of post-translational modifications

When analyzing clinical samples, researchers should consider using multiple complementary techniques and include appropriate controls to account for sample variability.

How does ATP5D expression differ across human tissues and pathological states?

ATP5D is expressed in multiple human tissues, reflecting the universal requirement for ATP synthesis. Notably, research has demonstrated significantly reduced ATP5D protein levels in the seminal plasma of infertile males compared to fertile controls . While ATP5D expression patterns in other pathological conditions are still being characterized, its altered expression in male infertility suggests it may serve as a potential biomarker. Researchers investigating tissue-specific expression should employ RNA-seq, proteomics, and tissue microarrays to establish comprehensive expression profiles in normal and disease states.

What is the molecular pathophysiology of ATP5D mutations in human disease?

Biallelic mutations in ATP5F1D cause a distinct metabolic disorder characterized by episodic lethargy, metabolic acidosis, 3-methylglutaconic aciduria, and hyperammonemia . The pathophysiology involves:

  • Impaired ATP synthase assembly: Mutations disrupt the proper assembly of the F1FO complex

  • Reduced Complex V activity: Leading to compromised ATP production capacity

  • Mitochondrial cristae abnormalities: Fibroblasts from affected individuals show significant decreases in mitochondrial cristae

  • Metabolic derangements: Including organic aciduria and hyperammonemia

Research approaches should include comprehensive metabolomics, mitochondrial functional assays, and electron microscopy to fully characterize the cascade of events resulting from ATP5D dysfunction.

What experimental models are most effective for studying ATP5D function and pathology?

Several experimental models have proven valuable for studying ATP5D:

  • Patient-derived fibroblasts: Demonstrate impaired ATP synthase assembly and cristae abnormalities

  • Drosophila models: Knockdown of ATPsynδ (the ATP5F1D homolog) causes developmental defects, with human wild-type ATP5F1D capable of rescuing the phenotype while mutant versions show partial rescue

  • Mouse models: ATP5D knockout and knockin models generated using CRISPR/Cas9 technology demonstrate fertility phenotypes

  • iPSC-derived models: Allow study of tissue-specific effects in relevant cell types

Each model offers distinct advantages: cell models allow biochemical analysis, Drosophila provides rapid generation time for genetic studies, while mouse models better recapitulate mammalian physiology.

How does ATP5D dysfunction contribute to male infertility?

Research has established a potential role for ATP5D in male fertility through multiple experimental approaches:

  • Expression correlation: ATP5D protein levels are significantly reduced in seminal plasma of infertile males

  • Knockout effects: ATP5D knockout male mice showed significantly prolonged fertility time and impaired testicular histomorphology

  • Functional implications: The involvement likely relates to the high energy requirements of spermatogenesis and sperm motility

Experimental approaches to investigate this association include ELISA measurement of ATP5D in seminal plasma, histological examination of testicular tissue, sperm functional assays, and metabolomic analysis of seminal fluid. Researchers should note that while expression levels are altered in infertility, sequencing studies did not detect mutations in the coding regions of ATP5D in infertile males .

What therapeutic strategies are being investigated for ATP5D-associated disorders?

While specific therapies for ATP5D-associated disorders are still emerging, several approaches warrant investigation:

  • Metabolic interventions: Management of metabolic acidosis and hyperammonemia

  • Mitochondrial biogenesis enhancers: Compounds that stimulate mitochondrial function

  • Gene therapy approaches: Viral vector-mediated delivery of functional ATP5F1D

  • Small molecule screening: Identification of compounds that might stabilize ATP synthase assembly

  • Precision medicine: Development of mutation-specific therapies for particular variants

Research methodologies should include high-throughput drug screening, patient-derived cell models for therapeutic testing, and appropriate animal models for preclinical evaluation.

How do ATP5D mutations specifically affect ATP synthase assembly and structure?

The p.Pro82Leu and p.Val106Gly mutations in ATP5F1D affect conserved residues that are critical for proper protein folding or interactions with other subunits . Functional studies have shown:

  • Cultured skin fibroblasts from affected individuals exhibit impaired assembly of F1FO ATP synthase

  • Subsequent reduction in complex V activity

  • Drosophila complementation studies demonstrate that mutant proteins retain partial function but cannot fully rescue developmental defects

Researchers investigating these effects should employ blue native PAGE, co-immunoprecipitation studies, cryo-electron microscopy, and molecular modeling to characterize structural perturbations caused by specific mutations.

What are the optimal approaches for investigating ATP5D interactions with other ATP synthase components?

To comprehensively characterize ATP5D interactions within the ATP synthase complex:

  • Blue native PAGE: Allows analysis of intact ATP synthase complexes

  • Co-immunoprecipitation: Identifies direct binding partners

  • Proximity labeling: BioID or APEX2 approaches can identify proteins in close proximity

  • Cryo-electron microscopy: Provides structural information about the assembled complex

  • Cross-linking mass spectrometry: Maps specific interaction interfaces

Researchers should combine multiple complementary approaches to build a comprehensive interaction map of ATP5D within the ATP synthase complex.

How can researchers effectively analyze ATP5D evolutionary conservation for functional insights?

ATP5D homologs are present across species, providing opportunities for evolutionary analysis:

  • The Drosophila homolog (ATPsynδ) functionally complements human ATP5F1D

  • Cross-species rescue experiments identify functionally conserved domains

  • Comparative genomics can highlight critical residues maintained throughout evolution

  • Phylogenetic analysis can reveal evolutionary relationships and functional constraints

Research approaches should combine computational phylogenetic analysis with experimental cross-species complementation to identify functionally critical domains and residues.

What considerations are important when establishing ATP5D as a biomarker for male fertility?

Research suggests ATP5D may serve as a potential biomarker for male fertility , requiring:

  • Standardized measurement protocols: Consistent ELISA or other quantitative methods

  • Large-scale validation: Studies across diverse populations to establish reference ranges

  • Correlation with clinical parameters: Sperm count, morphology, and other fertility metrics

  • Mechanism elucidation: Understanding how ATP5D affects spermatogenesis and sperm function

  • Comparative biomarker analysis: Determining ATP5D's value relative to existing fertility biomarkers

Researchers pursuing biomarker development should conduct prospective studies with appropriate statistical power and diverse patient populations to validate clinical utility.

What are the critical unanswered questions regarding ATP5D function in human disease?

Despite progress in understanding ATP5D, several important questions remain:

  • Tissue-specific effects: Why do ATP5D mutations predominantly affect specific tissues despite ubiquitous expression?

  • Phenotypic variability: What factors explain the clinical heterogeneity in patients with ATP5D mutations?

  • Regulatory mechanisms: How is ATP5D expression regulated across tissues and developmental stages?

  • Interaction partners: Are there tissue-specific interaction partners that modify ATP5D function?

  • Potential roles beyond ATP synthesis: Does ATP5D have functions independent of its role in ATP synthase?

These questions represent critical areas for future investigation that will advance our understanding of ATP5D biology and pathology.

How can systems biology approaches advance our understanding of ATP5D in cellular energetics?

Integrative approaches to study ATP5D include:

  • Multi-omics integration: Combining transcriptomics, proteomics, and metabolomics data

  • Network analysis: Identifying ATP5D within broader mitochondrial and cellular networks

  • Mathematical modeling: Creating predictive models of ATP synthase function incorporating ATP5D dynamics

  • Single-cell approaches: Characterizing cell-to-cell variability in ATP5D expression and function

  • In silico drug screening: Computational approaches to identify potential therapeutic compounds

These systems-level approaches can provide comprehensive insights into ATP5D's role within broader cellular energetic networks.

What are the recommended protocols for ATP5D functional analysis in patient samples?

For researchers working with patient samples to analyze ATP5D function:

  • ATP5D protein quantification: Validated ELISA assays for seminal plasma and other biofluids

  • Mitochondrial functional assessment: Oxygen consumption rate measurements in patient fibroblasts

  • ATP synthesis capacity: Luciferase-based ATP determination assays

  • Mitochondrial morphology: Electron microscopy protocols for cristae analysis

  • ATP synthase assembly: Blue native PAGE followed by immunoblotting for complex V subunits

Standardized protocols with appropriate controls are essential for meaningful cross-study comparisons and clinical correlations.

Product Science Overview

Introduction

ATP Synthase Subunit D, also known as ATP5PD, is a crucial component of the mitochondrial ATP synthase complex, often referred to as Complex V. This enzyme complex plays a pivotal role in cellular energy production by synthesizing adenosine triphosphate (ATP) from adenosine diphosphate (ADP) and inorganic phosphate, utilizing the proton gradient generated by the electron transport chain across the inner mitochondrial membrane .

Structure and Function

The mitochondrial ATP synthase is a multi-subunit protein complex with an approximate molecular weight of 550 kDa . It consists of two main functional domains: F1, located in the mitochondrial matrix, and Fo, embedded in the inner mitochondrial membrane . ATP Synthase Subunit D is part of the peripheral stalk, which connects these two domains and plays a critical role in the structural stability and function of the enzyme .

The peripheral stalk, including Subunit D, prevents the rotation of the catalytic F1 domain relative to the Fo domain during ATP synthesis. This stabilization is essential for the proper functioning of the rotary nanomotor mechanism of ATP synthase, which is responsible for the phosphorylation of ADP to ATP .

Genetic and Biochemical Aspects

The ATP5PD gene encodes the ATP Synthase Subunit D protein. Mutations in this gene can lead to various mitochondrial diseases, including Bardet-Biedl Syndrome 18 and other mitochondrial dysfunctions . The human recombinant form of this protein is often used in research to study its structure, function, and role in mitochondrial pathologies.

Role in Cellular Energy Production

ATP synthase is the smallest known biological nanomotor and is crucial for ATP production, which is the primary energy currency in all living cells . The enzyme operates through a process known as oxidative phosphorylation, where the energy from the proton gradient is harnessed to drive the synthesis of ATP. This process is vital for maintaining the energy balance within cells and supporting various metabolic pathways .

Pathological Implications

Deficiencies or malfunctions in ATP synthase, including Subunit D, can lead to severe mitochondrial diseases. These conditions often manifest as neuromuscular disorders, metabolic syndromes, and other systemic issues due to impaired cellular energy production . Understanding the structure and function of ATP Synthase Subunit D is therefore critical for developing therapeutic strategies for these diseases.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.