PTPMT1 Human

Protein Tyrosine Phosphatase, Mitochondrial 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

PTPMT1 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 199 amino acids (28-201) and having a molecular mass of 22.6kDa.
PTPMT1 is fused to a 25 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Protein tyrosine phosphatase mitochondrial 1 (PTPMT1) is a protein tyrosine phosphatase found in the inner membrane of mitochondria. It is highly expressed in various tissues, including pancreatic beta cells. PTPMT1 plays a crucial role in regulating mitochondrial function, particularly ATP production, by dephosphorylating specific mitochondrial proteins. It exhibits a preference for phosphatidylinositol 5-phosphate, a lipid signaling molecule, as a substrate.
Description
Recombinant PTPMT1, derived from humans and expressed in E. coli, is a non-glycosylated polypeptide consisting of 199 amino acids (residues 28-201). It has a molecular weight of 22.6 kDa. This PTPMT1 protein includes a 25 amino acid His-tag at the N-terminus to facilitate purification by proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile solution.
Formulation
PTPMT1 is supplied as a 1 mg/ml solution in a buffer consisting of 20mM Tris-HCl (pH 8.0), 10% glycerol, 1mM DTT, and 0.15M NaCl.
Stability
For short-term storage (up to 4 weeks), keep the PTPMT1 vial refrigerated at 4°C. For extended periods, store the protein frozen at -20°C. It is recommended to add a carrier protein like HSA or BSA (0.1%) for long-term storage. Avoid repeated freezing and thawing of the PTPMT1 solution.
Purity
The purity of PTPMT1 is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Protein-tyrosine phosphatase mitochondrial 1, PTEN-like phosphatase, Phosphoinositide lipid phosphatase, PTPMT1, MOSP, PLIP, DUSP23, PNAS-129.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMKVPGR AHRDWYHRID PTVLLGALPL RSLTRQLVQD ENVRGVITMN EEYETRFLCN SSQEWKRLGV EQLRLSTVDM TGIPTLDNLQ KGVQFALKYQ SLGQCVYVHC KAGRSRSATM VAAYLIQVHK WSPEEAVRAI AKIRSYIHIR PGQLDVLKEF HKQITARATK DGTFVISKT.

Q&A

What is PTPMT1 and what is its localization in human cells?

PTPMT1 is a highly conserved mitochondrial tyrosine phosphatase that localizes to the matrix leaflet of the inner mitochondrial membrane via an N-terminal signal sequence . It functions primarily as a phosphatase that dephosphorylates phosphatidylglycerol phosphate to phosphatidylglycerol, an essential intermediate within the cardiolipin biosynthetic pathway . Cardiolipin is the signature phospholipid of mitochondria and resides primarily in the inner mitochondrial membrane, where it plays fundamental roles in mitochondrial structure and function .

Methodologically, researchers can detect PTPMT1 localization through:

  • Subcellular fractionation followed by western blotting

  • Immunofluorescence microscopy with mitochondrial co-markers

  • Protease protection assays to confirm matrix-side orientation

What experimental models have been developed to study PTPMT1 function?

Several complementary experimental systems have been established to investigate PTPMT1:

  • Cellular models:

    • Patient-derived fibroblasts harboring PTPMT1 variants

    • Cellular rescue experiments with wild-type PTPMT1 expression

    • Cancer cell lines with PTPMT1 knockdown or inhibition

  • Animal models:

    • Global PTPMT1 knockout (embryonically lethal)

    • Tissue-specific knockout models:

      • Skeletal muscle-specific (PTPMT1fl/fl/CKMM-Cre+)

      • Cardiac-specific (PTPMT1fl/fl/MYH6-Cre+)

    • PTPMT1 knockout zebrafish

  • Biochemical assays:

    • Isolated mitochondria functional studies

    • Enzyme activity measurements

    • Lipidomic analyses of cardiolipin species

The complete ablation of PTPMT1 in mouse models results in embryonic lethality, highlighting its essential role in development .

How does PTPMT1 contribute to cardiolipin biosynthesis and what methods detect alterations?

PTPMT1 catalyzes a critical step in the de novo cardiolipin biosynthetic pathway by dephosphorylating phosphatidylglycerol phosphate to form phosphatidylglycerol . This intermediate is subsequently converted to cardiolipin, which is essential for mitochondrial function.

Researchers can detect cardiolipin alterations using:

  • Lipidomic analyses:

    • Liquid chromatography-mass spectrometry to quantify cardiolipin species

    • Reduced cardiolipin (72:8) levels were detected in muscle tissue and blood from individuals with PTPMT1 variants

  • Functional assays:

    • Respiratory chain enzyme analysis (e.g., Complex I activity measurements)

    • Mitochondrial membrane potential assessments

  • Clinical samples:

    • Dry blood spot analysis

    • Muscle tissue biopsies

Sample TypeTechniqueKey Finding in PTPMT1 Deficiency
Muscle tissueLC-MSReduced cardiolipin (72:8) levels
FibroblastsLipidomic analysisReduced total cardiolipin in cells with missense/splice variants
BloodDry blood spot analysisDetectable cardiolipin abnormalities

What is the relationship between cardiolipin abnormalities and mitochondrial morphology?

Loss of PTPMT1 function leads to decreased total cardiolipin content, which correlates with abnormal mitochondrial morphology . Research methodologies to assess this relationship include:

  • Microscopic techniques:

    • Electron microscopy to visualize ultrastructural changes

    • Fluorescence microscopy with mitochondrial-specific dyes

    • PTPMT1-deficient fibroblasts exhibit increased mitochondrial fragmentation

  • Quantitative analyses:

    • Computer-assisted morphometry of mitochondrial networks

    • Assessment of fusion/fission protein expression levels

  • Functional correlations:

    • Relationship between cardiolipin levels and respiratory complex assembly

    • Impact on mitochondrial membrane potential and ATP production

The observed mitochondrial fragmentation in PTPMT1-deficient cells can be rescued by exogenous expression of wild-type PTPMT1, confirming the causal relationship between PTPMT1 function, cardiolipin levels, and mitochondrial morphology .

What are the clinical manifestations of PTPMT1 variants in humans?

Biallelic PTPMT1 variants have been identified in six individuals from three unrelated families, presenting with a complex neurological and neurodevelopmental syndrome comprising :

  • Neurological features:

    • Developmental delay

    • Microcephaly

    • Epilepsy (onset at ~6 years in some patients)

    • Spasticity

    • Cerebellar ataxia and nystagmus

    • Optic atrophy

    • Bulbar dysfunction

  • Additional manifestations:

    • Facial dysmorphism

    • Sensorineural hearing loss

    • Hypoxic-ischaemic encephalopathy at birth (Subject S1)

    • Persistent pulmonary hypertension (Subject S1)

    • Hepatomegaly with elevated transaminases (Subject S1)

  • Neuroimaging findings:

    • Corpus callosum thinning

    • Cerebellar atrophy

    • White matter changes

    • Atrophy of pons and medulla in some cases

The disease severity appears to correlate with the degree of PTPMT1 protein loss, with minimal residual expression associated with milder phenotypes .

How are pathogenic PTPMT1 variants characterized at the molecular level?

Two types of pathogenic PTPMT1 variants have been identified and characterized through complementary approaches :

  • Variant identification:

    • Whole exome sequencing to identify candidate variants

    • Sanger sequencing for confirmation

    • Segregation analysis in affected families

  • Functional characterization:

    • mRNA analysis by qPCR:

      • Missense variant: normal transcript levels

      • Missense/splice region variant: severe reduction in transcript abundance (likely nonsense-mediated decay)

    • Protein analysis by western blot:

      • Decreased steady-state PTPMT1 protein levels in all patients

    • RNA-seq data to confirm transcript reduction

    • Complementation experiments with wild-type PTPMT1

  • In vivo modeling:

    • PTPMT1 knockout zebrafish showing similar phenotypes:

      • Abnormal body size

      • Developmental alterations

      • Decreased cardiolipin levels

      • OXPHOS deficiency

These analyses support a loss-of-function mechanism for PTPMT1 variants, affecting either protein stability or mRNA processing/stability .

How does PTPMT1 influence mitochondrial fuel selection and metabolism?

PTPMT1 plays a critical role in regulating mitochondrial substrate utilization, particularly facilitating the metabolism of carbohydrates . Research using tissue-specific knockout models has revealed several key mechanisms:

  • Altered substrate preference:

    • Decreased pyruvate utilization in PTPMT1-deficient mitochondria

    • Increased free fatty acid and glutamate utilization as compensatory mechanisms

  • Pyruvate metabolism defects:

    • 50% reduction in intramitochondrial pyruvate levels in PTPMT1-null mitochondria

    • Decreased α-ketoglutarate (α-KG) levels

    • Reduced acute production of α-KG from extramitochondrial pyruvate

    • Normal pyruvate dehydrogenase (PDH) activity

  • Metabolic adaptations:

    • Muscle fiber-type switching from oxidative to glycolytic fibers

    • Dramatic increase (~170-fold) in MYH4 (marker of glycolytic fast-twitch Type 2B fibers)

These findings suggest that PTPMT1 specifically affects pyruvate uptake into mitochondria, rather than its subsequent metabolism, potentially through modulation of mitochondrial membrane properties or transporter functions .

What experimental approaches are used to assess mitochondrial function in PTPMT1-deficient tissues?

Researchers employ multiple complementary techniques to evaluate mitochondrial dysfunction in PTPMT1-deficient models:

  • Bioenergetic assessments:

    • Respiratory chain enzyme analysis (specific activity measurements)

    • Complex I activity reduction (~50%) in muscle tissue from PTPMT1-deficient individuals

    • ATP level measurements in tissues and isolated mitochondria

  • Metabolic substrate utilization:

    • Oxygen consumption measurements with different substrates

    • Direct measurement of metabolite levels (pyruvate, α-KG, acetyl-CoA)

    • Acute metabolite production from exogenous substrates

  • Signaling pathway analysis:

    • Assessment of energy stress markers (p-AMPK, p-mTOR)

    • 10-12 month-old PTPMT1fl/fl/MYH6-Cre+ hearts show increased p-AMPK (T172) and decreased p-mTOR (S2448)

  • Mitochondrial content and integrity:

    • Mitochondrial DNA quantification

    • Expression analysis of mitochondrial complexes

    • Membrane potential measurements (JC-1 staining)

These approaches have revealed tissue-specific and age-dependent consequences of PTPMT1 deficiency, with cardiac dysfunction becoming apparent only in older knockout mice despite early metabolic alterations .

What is the potential role of PTPMT1 in cancer biology and therapeutics?

PTPMT1 has emerged as a potential therapeutic target in cancer, particularly in small cell lung cancer (SCLC) :

  • Expression analysis in cancer:

    • Immunohistochemical staining of SCLC tissues

    • Western blot and qRT-PCR quantification in cancer cell lines versus normal tissues

  • Functional studies:

    • PTPMT1 knockdown using lentivirus-mediated shRNA transduction

    • Pharmacological inhibition with alexidine dihydrochloride

    • Effects on cell viability (CCK-8 assay)

    • Colony formation capacity

    • Cell migration properties

  • Mechanism investigations:

    • Mitochondrial function assessment (JC-1 staining)

    • Transcriptome sequencing

    • Untargeted metabolomics analysis

These studies have demonstrated that PTPMT1 inhibition induces apoptosis and growth arrest in SCLC cells, suggesting its critical role in cancer cell survival and growth . The development of selective PTPMT1 inhibitors may represent a novel therapeutic approach for SCLC treatment.

How do researchers differentiate between PTPMT1's roles in phosphoinositide metabolism versus cardiolipin biosynthesis?

PTPMT1 has dual functions in mitochondria - dephosphorylating phosphatidylinositol phosphates (PIPs) and participating in cardiolipin biosynthesis . Distinguishing between these functions requires sophisticated experimental approaches:

  • Substrate-specific assays:

    • In vitro enzymatic assays with purified PTPMT1 and different substrates

    • Phosphatase activity measurements toward PIPs versus phosphatidylglycerol phosphate

  • Rescue experiments with mutants:

    • Structure-function analysis with PTPMT1 variants affecting specific catalytic activities

    • Complementation with domain-specific mutants to separate functions

  • Metabolite profiling:

    • Comprehensive lipidomic analysis of both PIPs and cardiolipin species

    • Correlation between specific lipid alterations and phenotypic consequences

  • Downstream effector analysis:

    • Assessment of UCP2 activation (linked to PIP metabolism)

    • Mitochondrial uncoupling measurements

    • C4 dicarboxylate intermediate transport

Research suggests that PTPMT1 facilitates mitochondrial metabolism largely through dephosphorylation of PIP substrates that appear to inhibit mitochondrial oxidative phosphorylation but enhance cytosolic glycolysis by activating mitochondrial uncoupling protein 2 (UCP2) .

What is the relationship between PTPMT1 and other cardiolipin-related disease genes?

PTPMT1 has been established as a new cardiolipin-related primary mitochondrial disease (PMD) gene, contributing to the growing spectrum of Mendelian disorders associated with aberrant cardiolipin metabolism :

  • Comparative phenotype analysis:

    • TAZ (Barth syndrome): severe myopathy, cardiomyopathy, neutropenia

    • PNPLA8, CRLS1, TAMM41, PTPMT1: multisystemic disorders with progressive encephalopathy and neurodevelopmental regression

  • Tissue-specificity investigations:

    • Despite cardiolipin abundance in heart tissue, cardiac manifestations were not prominent in PTPMT1-deficient individuals

    • Similar findings with other cardiolipin-related genes (e.g., TAMM41)

  • Cardiolipin biology research:

    • Factors regulating tissue-specific acyl chain composition of cardiolipin

    • Contribution of different cardiolipin-related genes to human pathophysiology

The emerging picture suggests complex relationships between specific cardiolipin metabolic defects and clinical manifestations, with ongoing research needed to understand the variable clinical phenotypes linked to abnormal cardiolipin species .

What methodological approaches can improve detection and functional characterization of PTPMT1 variants?

Advanced techniques for identifying and characterizing PTPMT1 variants include:

  • Enhanced genetic screening:

    • Inclusion of PTPMT1 in gene panels for mitochondrial disorders

    • RNA sequencing to detect splicing abnormalities

    • Long-read sequencing to identify structural variants

  • Functional genomics:

    • CRISPR-Cas9 engineering of specific variants in cell models

    • High-throughput screening of variant effects on PTPMT1 function

    • iPSC-derived organoids to model tissue-specific effects

  • Structure-based analyses:

    • Protein structure modeling to predict variant impact

    • In silico simulation of variant effects on PTPMT1 catalytic activity

    • Drug screening for variant-specific chaperones or activators

  • Novel biomarkers:

    • Development of specific cardiolipin profiles as diagnostic markers

    • Correlation of cardiolipin species with disease severity

    • Non-invasive detection methods for routine clinical implementation

These approaches can facilitate earlier diagnosis and potentially guide personalized therapeutic strategies for individuals with PTPMT1-related disorders.

Product Science Overview

Structure and Function

PTPMT1 is a mitochondrial protein that specifically mediates the dephosphorylation of mitochondrial proteins, thereby playing an essential role in ATP production . It has been shown to display phosphatase activity toward phosphoprotein substrates, which is critical for maintaining cellular energy homeostasis .

Biological Significance

PTPMT1 is involved in several metabolic pathways, including glycerophospholipid biosynthesis and general metabolism . Its activity is essential for the proper functioning of mitochondrial processes, which are vital for energy production and overall cellular health .

Expression Patterns and Tissue Distribution

PTPMT1 is ubiquitously expressed in various tissues, with significant roles in lymphoid tissue, bone marrow, testis, and skeletal muscle . It is also involved in immune responses and protein ubiquitination, highlighting its diverse functional roles in different biological contexts .

Clinical Relevance

Mutations or dysregulation of PTPMT1 have been associated with several diseases, including primary ciliary dyskinesia and Barth syndrome . Given its central role in mitochondrial function and energy production, PTPMT1 is a potential target for therapeutic interventions aimed at treating metabolic disorders and mitochondrial diseases .

Research and Therapeutic Potential

Ongoing research is focused on understanding the detailed mechanisms of PTPMT1’s action and its interactions with other proteins. This knowledge is crucial for developing specific inhibitors or modulators that can be used in clinical settings to treat diseases associated with mitochondrial dysfunction .

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.