ATP5H Human

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

Description

Functional Role in Mitochondrial ATP Synthase

ATP5H is integral to the F₀ stator stalk, which:

  1. Stabilizes the F₀-F₁ interface during rotational catalysis

  2. Facilitates proton translocation across the inner mitochondrial membrane

  3. Maintains structural integrity of ATP synthase’s peripheral stalk

Disruption of ATP5H impairs:

  • ATP production (ΔΨm\Delta \Psi_m reduction)

  • Oxygen consumption rates

  • Mitochondrial membrane potential

Cancer

FindingMechanismClinical ImpactSource
ATP5H lossHDAC1-mediated histone deacetylationMultimodal therapy resistance (immunotherapy/chemotherapy/radiotherapy)
ROS accumulationHIF-1α stabilization → AKT/ERK activationTumor immune evasion

Neurodegeneration

  • Alzheimer’s disease:

    • rs11870474 SNP (OR = 1.53, P=4.7×109P = 4.7 \times 10^{-9}) at ATP5H/KCTD2 locus

    • Impaired neuronal hyperpolarization under hypoxia

Recombinant ATP5H Production

Applications:

  • Mechanistic studies of oxidative phosphorylation

  • Antibody production

  • In vitro models of mitochondrial dysfunction

Research Advancements

Key discoveries from recent studies:

  1. Immune editing: Tumor cells epigenetically silence ATP5H via HDAC1 upregulation, reducing histone H4 acetylation at promoter regions (P<0.01P < 0.01) .

  2. Therapeutic resistance:

    • ATP5H knockdown increases glucose uptake (+38%) and lactate production (+52%)

    • Restoring ATP5H expression resensitizes P3 cancer cells to therapy (P<0.05P < 0.05)

Product Specs

Introduction
ATP5H, a member of the ATPase d subunit family, encodes the d subunit of the F0 complex. This complex plays a crucial role in ATP synthesis within the mitochondria. Specifically, electron transport complexes in the respiratory chain generate a proton gradient across the membrane, which drives ATP5H to convert ADP to ATP. Located on human chromosome 17q25.1, the ATP5H gene encodes two alternatively spliced isoforms. Notably, ATP5H is exclusively found within the mitochondrial inner membrane.
Description
Recombinant ATP5H Human, produced in E.coli, is a single, non-glycosylated polypeptide chain consisting of 184 amino acids (residues 1-161). It has a molecular weight of 20.9 kDa. For purification purposes, a 23 amino acid His-tag is fused to the N-terminus of ATP5H. The protein is then purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The ATP5H solution is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein such as 0.1% HSA or BSA is advisable for long-term storage. Avoid repeated freezing and thawing of the solution.
Purity
The purity of the protein is greater than 85% as determined by SDS-PAGE analysis.
Synonyms
ATP Synthase H+ Transporting, Mitochondrial Fo Complex Subunit D, ATP Synthase D Chain Mitochondrial, ATP Synthase H+ Transporting Mitochondrial F1F0 Subunit D, ATPase Subunit D, My032 Protein, ATPQ.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAGRKLA LKTIDWVAFA EIIPQNQKAI ASSLKSWNET LTSRLAALPE NPPAIDWAYY KANVAKAGLV DDFEKKFNAL KVPVPEDKYT AQVDAEEKED VKSCAEWVSL SKARIVEYEK EMEKMKNLIP FDQMTIEDLN EAFPETKLDK KKYPYWPHQP IENL

Q&A

What is ATP5H and what is its primary function in human cells?

ATP5H, also known as ATP5PD (ATP synthase peripheral stalk subunit d), is a critical component of mitochondrial ATP synthase. It specifically encodes subunit d of the peripheral stalk in the F0 complex of the enzyme. Mitochondrial ATP synthase catalyzes ATP synthesis by utilizing an electrochemical gradient of protons across the inner mitochondrial membrane during oxidative phosphorylation. The enzyme consists of two linked multi-subunit complexes: the soluble catalytic core (F1) and the membrane-spanning component (F0), with ATP5H being part of the F0 complex that comprises the proton channel . The proper functioning of ATP5H is essential for efficient energy production in eukaryotic cells, making it a fundamental component of cellular bioenergetics and metabolism.

How is ATP5H gene expression regulated in normal human tissues?

ATP5H gene expression is regulated through several mechanisms, with epigenetic control playing a particularly important role. Under normal conditions, the ATP5H promoter region maintains an appropriate level of histone acetylation, particularly H4 acetylation, which facilitates gene transcription. Research demonstrates that histone deacetylases (HDACs), especially HDAC1, can significantly impact ATP5H expression by modifying the acetylation status of histones at the ATP5H promoter region . Additionally, alternative splicing mechanisms generate different transcript variants encoding distinct isoforms of ATP5H. Researchers investigating ATP5H regulation should employ chromatin immunoprecipitation (ChIP) assays to assess histone modifications at the promoter region, RNA sequencing to identify transcript variants, and protein analysis methods to measure resulting protein levels in different tissue types.

What are the structural characteristics of the ATP5H protein?

The ATP5H protein (ATP synthase subunit d) is a component of the peripheral stalk of mitochondrial ATP synthase. It belongs to the PF05873 Pfam protein family . The protein contributes to the structural stability of the ATP synthase complex, particularly in maintaining the integrity of the peripheral stalk that connects the F1 and F0 components. Researchers studying ATP5H structure should employ X-ray crystallography or cryo-electron microscopy to determine its three-dimensional conformation within the larger ATP synthase complex. Additionally, protein-protein interaction studies using techniques such as co-immunoprecipitation or proximity ligation assays can help elucidate how ATP5H interacts with other subunits of the ATP synthase complex, providing insights into its functional role within the enzyme assembly.

How does ATP5H loss affect cancer cell metabolism?

ATP5H loss triggers profound mitochondrial metabolic reprogramming in cancer cells. When ATP5H expression is downregulated, cancer cells exhibit several key metabolic alterations: (1) elevated mitochondrial membrane potential, (2) defective ATP production, (3) reduced oxygen consumption, (4) enhanced glucose uptake, and (5) increased lactate production . This metabolic shift represents a classic Warburg effect phenotype, where cells increasingly rely on glycolysis even in the presence of oxygen. Methodologically, researchers should assess these metabolic parameters using techniques such as Seahorse XF analysis for oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), ATP luminescence assays, glucose uptake measurements using fluorescent glucose analogs, and lactate production assays. Importantly, these metabolic alterations occur without significant AMPK activation or spontaneous autophagy induction, suggesting that ATP5H loss does not exert significant metabolic stress on tumor cells despite the major bioenergetic reprogramming .

What molecular mechanisms link ATP5H loss to therapy resistance in cancer?

ATP5H loss promotes multimodal resistance to therapy through a linear pathway involving ROS accumulation and HIF-1α stabilization. The molecular pathway proceeds as follows: (1) ATP5H downregulation causes mitochondrial dysfunction leading to increased ROS production; (2) elevated ROS levels stabilize HIF-1α protein under normoxic conditions; (3) stabilized HIF-1α activates the AKT/ERK signaling pathway; (4) this signaling cascade ultimately confers resistance to immunotherapy, chemotherapy, and radiotherapy . To investigate this pathway, researchers should employ siRNA knockdown of ATP5H followed by ROS measurements using fluorescent probes such as DCFDA, Western blotting for HIF-1α protein levels and phosphorylated AKT/ERK, and cell viability assays after exposure to various therapeutic agents. Validation experiments should include treatment with antioxidants (such as N-acetyl cysteine or MitoTempo) and HIF-1α inhibitors to confirm the causal relationship between these molecular events and therapy resistance.

How does immune editing affect ATP5H expression in tumor cells?

Immune editing triggers epigenetic silencing of ATP5H through histone deacetylation. During sequential rounds of immune selection via cytotoxic T lymphocytes, tumor cells gradually lose ATP5H expression at the transcriptional level . This process is mediated by HDAC1 upregulation, which causes deacetylation of histone H4 at the ATP5H promoter region. Methodologically, researchers can study this phenomenon using a sequential immune editing model where tumor cells are repeatedly exposed to tumor-specific cytotoxic T lymphocytes. ATP5H expression should be monitored via qRT-PCR and Western blotting after each round of selection. Chromatin immunoprecipitation (ChIP) assays pulling down histone H3 and H4 followed by qPCR for ATP5H promoter sequences can reveal changes in histone acetylation status. Treatment with HDAC inhibitors (such as trichostatin A) or HDAC1-specific siRNA can confirm the role of histone deacetylation in ATP5H silencing. Importantly, DNA methylation inhibitors have been shown not to affect ATP5H expression, suggesting that histone deacetylation, rather than DNA methylation, is the primary epigenetic mechanism regulating ATP5H in this context .

What are the optimal methods for manipulating ATP5H expression in experimental models?

For effective ATP5H expression manipulation, researchers should consider several approaches based on experimental goals. For transient knockdown, siRNA transfection targeting ATP5H has been demonstrated to effectively reduce expression. The research literature shows successful ATP5H knockdown using this approach, with phenotypic changes visible within 48-72 hours post-transfection . For stable knockdown, shRNA delivered via lentiviral vectors provides longer-term suppression. For overexpression studies, transfection with expression vectors containing ATP5H cDNA under a strong promoter has been shown to restore ATP5H levels in deficient cells .

To validate the specificity of ATP5H manipulation, researchers should:

  • Confirm expression changes at both mRNA (qRT-PCR) and protein (Western blot) levels

  • Include appropriate controls (scrambled siRNA for knockdown studies)

  • Assess functional outcomes such as ATP production, oxygen consumption, and mitochondrial membrane potential

  • Consider rescue experiments by re-expressing ATP5H in knockdown models to demonstrate phenotype reversal

When studying ATP5H in the context of immune editing, the sequential exposure of tumor cells to cytotoxic T lymphocytes provides a valuable model system that recapitulates the gradual loss of ATP5H observed in therapy-resistant cancers .

What techniques are most effective for measuring the functional consequences of ATP5H alterations?

To comprehensively assess the functional impact of ATP5H alterations, researchers should employ a multiparametric approach targeting key mitochondrial and cellular functions:

  • Mitochondrial Function Assessment:

    • Measure mitochondrial membrane potential using fluorescent dyes such as JC-1 or TMRM

    • Quantify ATP production using luminescence-based ATP assays

    • Analyze oxygen consumption rate (OCR) using Seahorse XF analyzer

    • Assess glucose uptake with fluorescent glucose analogs (2-NBDG)

    • Measure lactate production using enzymatic assays

  • ROS Measurement:

    • Quantify cellular ROS using DCFDA or other fluorescent ROS indicators

    • Employ mitochondria-specific ROS probes such as MitoSOX

    • Validate ROS involvement using antioxidants (NAC) or mitochondria-targeted antioxidants (MitoTempo)

  • Therapeutic Response Testing:

    • Evaluate sensitivity to immunotherapy using CTL-mediated killing assays

    • Assess chemosensitivity with standard cytotoxicity assays

    • Determine radioresistance using clonogenic survival assays after irradiation

  • Signaling Pathway Analysis:

    • Measure HIF-1α protein levels via Western blotting

    • Quantify phosphorylation of AKT and ERK

    • Assess downstream targets such as VEGF secretion using ELISA

This comprehensive approach enables researchers to establish causal relationships between ATP5H status and cellular phenotypes, validating experimental findings through multiple complementary techniques.

How can researchers effectively model ATP5H loss in in vivo systems?

Developing effective in vivo models to study ATP5H loss requires careful consideration of multiple approaches:

  • Genetic Manipulation Strategies:

    • Conditional knockout models using Cre-loxP systems provide tissue-specific ATP5H deletion

    • CRISPR/Cas9-mediated genome editing can generate precise mutations or deletions

    • Xenograft models using ATP5H-silenced cancer cells enable assessment of tumor growth and therapy response in vivo

  • Immune Editing Models:

    • Sequential in vivo passaging of tumor cells under immune selection pressure can recapitulate the gradual loss of ATP5H observed in therapy-resistant cancers

    • Adoptive transfer of tumor-specific CTLs provides a defined immune selection mechanism

  • Therapeutic Intervention Approaches:

    • Administer antioxidants (e.g., NAC) to reverse phenotypic changes associated with ATP5H loss

    • Test HDAC inhibitors to prevent or reverse ATP5H silencing

    • Evaluate combination therapies targeting both ATP5H-loss phenotypes and conventional treatment modalities

  • Assessment Parameters:

    • Monitor tumor growth kinetics

    • Evaluate therapy responses (immunotherapy, chemotherapy, radiotherapy)

    • Analyze tumor tissue for ATP5H expression, ROS levels, HIF-1α stabilization, and AKT/ERK activation

    • Assess metastatic potential and invasiveness

For effective translation to human disease, researchers should validate findings from mouse models by analyzing ATP5H expression in patient tumor samples and correlating with clinical outcomes, therapy responses, and disease progression .

How does ATP5H expression correlate with clinical outcomes in cancer patients?

ATP5H loss in tumors has been strongly linked to therapy failure, disease progression, and poor survival in cancer patients . This correlation highlights the potential clinical significance of ATP5H as a biomarker for treatment response and prognosis. To investigate this relationship, researchers should employ the following methodological approaches:

This comprehensive analysis can establish ATP5H as a clinically relevant biomarker and potential therapeutic target in cancer management.

What therapeutic strategies might counteract the effects of ATP5H loss in cancer?

Based on the mechanistic understanding of ATP5H loss-induced phenotypes, several therapeutic strategies could potentially counteract its effects:

  • Antioxidant Therapy:

    • Administration of antioxidants such as N-acetyl cysteine (NAC) has been demonstrated to reduce ROS levels in ATP5H-deficient cells and restore their sensitivity to immunotherapy, chemotherapy, and radiotherapy

    • Mitochondria-targeted antioxidants like MitoTempo show similar effects and may provide more specific targeting of the mitochondrial ROS that drives the resistance phenotype

  • Epigenetic Modulation:

    • HDAC inhibitors, particularly those targeting HDAC1, can potentially reverse the epigenetic silencing of ATP5H

    • Treatment with trichostatin A (TSA) has been shown to upregulate ATP5H transcription in immune-edited tumor cells

  • HIF-1α Pathway Inhibitors:

    • Since HIF-1α stabilization is a key downstream effect of ATP5H loss, HIF-1α inhibitors could potentially reverse the resistance phenotype

    • AKT/ERK pathway inhibitors might also counteract the signaling changes induced by ATP5H loss

  • Metabolic Targeting:

    • Exploiting the altered metabolic state of ATP5H-deficient cells through glucose uptake inhibitors or glycolysis modulators

For clinical implementation, researchers should design rational combination strategies that pair these ATP5H-targeted approaches with conventional therapies to overcome resistance and improve patient outcomes.

How does ATP5H loss impact the tumor microenvironment and immune surveillance?

ATP5H loss in tumor cells may significantly alter the tumor microenvironment and immune surveillance through multiple mechanisms:

  • Metabolic Competition:

    • The increased glucose uptake and glycolytic metabolism in ATP5H-deficient tumor cells may create a glucose-depleted microenvironment

    • This metabolic competition could impair T cell function, as activated T cells also rely heavily on glycolysis

  • HIF-1α-Mediated Effects:

    • HIF-1α stabilization in ATP5H-deficient cells leads to increased VEGF secretion , potentially promoting angiogenesis and altering the vascular architecture of tumors

    • HIF-1α activation may also upregulate immune checkpoint molecules like PD-L1, contributing to immune evasion

  • ROS-Mediated Signaling:

    • Elevated ROS production by ATP5H-deficient tumor cells may affect adjacent stromal and immune cells through paracrine signaling

    • ROS can modulate the function of tumor-infiltrating lymphocytes and myeloid-derived suppressor cells

  • Invasive Phenotype Consequences:

    • The stem-like and invasive phenotype acquired by ATP5H-deficient cells may affect their interaction with extracellular matrix components and stromal cells

To investigate these complex interactions, researchers should employ:

  • Co-culture systems with tumor cells and immune components

  • Multiplex immunohistochemistry to assess the immune infiltrate in ATP5H-high versus ATP5H-low tumors

  • Single-cell RNA sequencing to characterize the heterogeneity of tumor and immune cells

  • Metabolic profiling of the tumor microenvironment using imaging mass spectrometry

These approaches can provide insights into how ATP5H status in tumor cells shapes the broader tumor ecosystem and influences response to immunotherapy.

What is the relationship between ATP5H function and mitochondrial dynamics in cellular stress responses?

The relationship between ATP5H function and mitochondrial dynamics during cellular stress represents an important frontier in understanding mitochondrial biology:

Research methodologies to explore these questions should include:

  • Live-cell imaging of mitochondrial networks using fluorescent markers

  • Electron microscopy to assess ultrastructural changes

  • Protein interaction studies focused on ATP5H and components of mitochondrial dynamics machinery

  • Assessment of mitophagy flux in ATP5H-manipulated cells

  • Analysis of mitochondrial DNA integrity and copy number

Understanding this relationship could reveal new therapeutic opportunities targeting mitochondrial dynamics in ATP5H-deficient cancer cells.

How does ATP5H interact with other components of the ATP synthase complex to maintain mitochondrial function?

Understanding the structural and functional interactions of ATP5H within the ATP synthase complex remains an important research question:

  • Structural Integration:

    • ATP5H (subunit d) is part of the peripheral stalk of ATP synthase, which connects the F1 catalytic domain to the membrane-embedded F0 domain

    • The precise positioning of ATP5H within this structure and its interactions with other peripheral stalk components (such as subunits b, F6, and OSCP) warrant detailed investigation

  • Assembly Dynamics:

    • The role of ATP5H in the stepwise assembly of the ATP synthase complex remains incompletely understood

    • Whether ATP5H loss affects the incorporation of other subunits or triggers compensatory changes in complex composition requires further study

  • Functional Coupling:

    • How ATP5H contributes to the mechanical coupling between proton translocation and ATP synthesis

    • Whether ATP5H participates in the regulation of ATP synthase activity in response to cellular energy demands

  • Supramolecular Organization:

    • ATP synthase forms dimers and oligomers that shape the inner mitochondrial membrane

    • ATP5H may contribute to these higher-order structures, influencing both enzyme function and mitochondrial morphology

Research approaches to address these questions include:

  • Cryo-electron microscopy of intact ATP synthase complexes with and without ATP5H

  • Cross-linking mass spectrometry to map protein-protein interaction surfaces

  • Site-directed mutagenesis of ATP5H followed by functional analysis

  • In vitro reconstitution of ATP synthase with modified or absent ATP5H

These studies would provide fundamental insights into mitochondrial bioenergetics and potentially reveal new approaches to modulate ATP synthase function in disease states.

Table 1: Phenotypic Changes Associated with ATP5H Loss in Cancer Cells

ParameterNormal ATP5H ExpressionATP5H LossMethod of AssessmentReference
Mitochondrial Membrane PotentialNormalElevatedJC-1 or TMRM fluorescence
ATP ProductionNormalDecreasedLuminescence-based ATP assay
Oxygen ConsumptionNormalReducedSeahorse XF analyzer
Glucose UptakeNormalEnhancedFluorescent glucose analog uptake
Lactate ProductionNormalIncreasedEnzymatic assay
ROS LevelsLowElevatedDCFDA fluorescence
HIF-1α Protein LevelLow (degraded)High (stabilized)Western blot
AKT/ERK PhosphorylationBaselineIncreasedWestern blot
VEGF SecretionBaselineIncreasedELISA
Response to ImmunotherapySensitiveResistantCTL killing assay
Response to ChemotherapySensitiveResistantCytotoxicity assay
Response to RadiotherapySensitiveResistantClonogenic survival assay

Table 2: Therapeutic Interventions That Reverse ATP5H Loss-Induced Phenotypes

InterventionTarget MechanismEffect on ATP5H-Deficient CellsMethod of AssessmentReference
N-acetyl cysteine (NAC)ROS scavengingReduces ROS, restores therapy sensitivityDCFDA fluorescence, therapy response assays
MitoTempoMitochondrial ROS scavengingReduces ROS, restores therapy sensitivityDCFDA fluorescence, therapy response assays
Trichostatin A (TSA)HDAC inhibitionRestores ATP5H expressionqRT-PCR, Western blot
HDAC1 siRNAHDAC1 knockdownIncreases ATP5H expression, enhances H4 acetylationqRT-PCR, Western blot, ChIP
ATP5H overexpressionDirect restorationNormalizes mitochondrial function, reduces ROS, restores therapy sensitivityMultiple functional assays
HIF-1α knockdownHIF-1α reductionReverses AKT/ERK activation and VEGF secretionWestern blot, ELISA

What emerging technologies could advance our understanding of ATP5H biology?

Future research on ATP5H biology could be significantly enhanced by several emerging technologies:

  • CRISPR Screening Approaches:

    • Genome-wide CRISPR screens in the context of ATP5H deficiency could identify synthetic lethal interactions

    • CRISPRi/CRISPRa systems enable fine-tuned modulation of ATP5H expression

  • Single-Cell Technologies:

    • Single-cell RNA sequencing could reveal heterogeneity in ATP5H expression within tumors

    • Single-cell metabolomics may uncover cell-specific metabolic reprogramming following ATP5H loss

  • Advanced Imaging Techniques:

    • Super-resolution microscopy can visualize ATP5H localization within the ATP synthase complex

    • Live-cell imaging with genetically encoded biosensors can monitor real-time changes in ATP, ROS, and mitochondrial dynamics

  • Structural Biology Innovations:

    • Cryo-electron tomography of intact mitochondria can examine ATP synthase in its native membrane environment

    • Integrative structural approaches combining NMR, X-ray crystallography, and computational modeling

  • Proteomics and Interactomics:

    • Proximity labeling techniques like BioID or APEX can map the ATP5H interactome

    • Phosphoproteomics can identify signaling changes downstream of ATP5H loss

These technological advances will provide unprecedented insights into ATP5H function and potentially reveal new therapeutic approaches for diseases associated with ATP5H dysregulation.

How might ATP5H research influence our understanding of other mitochondrial diseases?

ATP5H research has broader implications for understanding mitochondrial diseases:

  • Mechanistic Parallels:

    • The ROS-dependent signaling observed in ATP5H-deficient cells may represent a common pathway in various mitochondrial disorders

    • The epigenetic regulation of ATP5H might inform similar mechanisms affecting other mitochondrial genes

  • Therapeutic Translation:

    • Antioxidant approaches effective in ATP5H-deficient models could benefit other mitochondrial diseases characterized by oxidative stress

    • Metabolic interventions targeting the altered bioenergetic state might have broad applicability

  • Biomarker Development:

    • ATP5H expression or functional status could serve as a biomarker for mitochondrial dysfunction in various pathological conditions

    • The downstream effects of ATP5H loss (HIF-1α stabilization, AKT/ERK activation) might represent more accessible biomarkers

  • Evolutionary Conservation:

    • Comparative studies of ATP5H across species could reveal fundamental aspects of mitochondrial evolution and adaptation

    • Conservation of ATP5H regulation mechanisms might highlight evolutionary pressure points in mitochondrial function

By integrating findings from ATP5H research with broader studies of mitochondrial biology, researchers can develop more comprehensive models of mitochondrial dysfunction in disease and identify common therapeutic targets across multiple conditions.

Product Science Overview

Introduction

ATP synthase is a crucial enzyme in cellular energy production, responsible for synthesizing adenosine triphosphate (ATP), the primary energy currency of the cell. The enzyme is located in the inner mitochondrial membrane and is composed of multiple subunits, forming a complex known as the F1Fo ATP synthase or Complex V. Among these subunits, the Fo complex subunit D plays a significant role in the enzyme’s function.

Structure and Function

The ATP synthase enzyme consists of two main components: the F1 and Fo complexes. The F1 complex is situated in the mitochondrial matrix, while the Fo complex is embedded in the inner mitochondrial membrane. The Fo complex forms a channel through which protons flow, driving the rotation of the F1 complex and facilitating ATP synthesis from adenosine diphosphate (ADP) and inorganic phosphate (Pi) .

The Fo complex subunit D, also known as ATP5PD, is a peripheral stalk subunit that helps stabilize the interaction between the F1 and Fo complexes. This stabilization is essential for the proper functioning of the ATP synthase enzyme, ensuring efficient ATP production .

Role in Oxidative Phosphorylation

ATP synthase is a key player in oxidative phosphorylation, the process by which cells generate ATP through the electron transport chain. During oxidative phosphorylation, electrons are transferred through a series of protein complexes in the inner mitochondrial membrane, creating a proton gradient. The energy from this gradient is harnessed by ATP synthase to produce ATP .

Human Recombinant ATP Synthase Fo Complex Subunit D

Recombinant DNA technology allows for the production of human proteins in various host systems, such as bacteria, yeast, or mammalian cells. Human recombinant ATP synthase Fo complex subunit D is produced by inserting the gene encoding this subunit into a host organism, which then expresses the protein. This recombinant protein can be used for research purposes, such as studying the structure and function of ATP synthase, as well as for potential therapeutic applications.

Pathological Implications

Mutations or deficiencies in ATP synthase subunits, including the Fo complex subunit D, can lead to mitochondrial diseases. These diseases often result in impaired energy production, leading to a range of symptoms, including muscle weakness, neurological disorders, and metabolic abnormalities. Understanding the structure and function of ATP synthase subunits is crucial for developing potential treatments for these conditions .

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.