PPM1F Antibody

Protein Phosphatase 1F, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Applications in Research

PPM1F antibodies are widely used to investigate integrin regulation, cancer biology, and neurological disorders. Key applications include:

ApplicationKey FindingsReferences
Western Blot (WB)Detects endogenous PPM1F (~50 kDa) in glioblastoma, fibroblasts, and platelets.
ImmunofluorescenceLocalizes active integrin β1 and talin clusters in focal adhesions.
Knockdown/KO StudiesValidates PPM1F depletion in CRISPR/Cas9-KO cells (e.g., A172 glioblastoma).
Disease ModelsLinks PPM1F to depression (via AMPK signaling) and cancer progression.

Integrin Regulation

PPM1F dephosphorylates the integrin β1 T788/T789 motif, acting as a phospho-switch to:

  • Suppress integrin activity: Dephosphorylation dissociates filaminA and recruits talin/kindlin-2, modulating cell adhesion .

  • Enhance adhesion in KO models: PPM1F-deficient cells exhibit 1.5–2× increased adhesion to extracellular matrix ligands .

Neurological and Cancer Roles

  • Depression: Reduced PPM1F in the medial prefrontal cortex (mPFC) activates AMPK, inducing microglial inflammation and depressive behaviors in mice .

  • Cancer Biomarker: High PPM1F correlates with poor survival in hepatocellular carcinoma (HCC) and ovarian cancer .

Validation and Quality Control

  • Specificity: Validated via KO cell lines (e.g., A172 PPM1F-KO shows no off-target effects on focal adhesion proteins) .

  • Dilution Range: WB: 1:5,000–1:50,000; IHC: 1:100–1:500 .

Product Specs

Introduction
Protein Phosphatase 1F (PPM1F), a member of the PP2C family of Ser/Thr protein phosphatases, acts as a negative regulator of cell stress response pathways. PPM1F interacts with Rho guanine nucleotide exchange factors (PIX), thereby inhibiting the activity of p21-activated kinase 1 (PAK), a protein kinase involved in mediating the downstream effects of Rho GTPases. Notably, PPM1F dephosphorylates Calcium/calmodulin-dependent protein kinase II gamma (CAMK2G/CAMK-II) and has been identified as a promoter of apoptosis.
Physical Appearance
Sterile, colorless solution.
Formulation
The antibody is supplied at a concentration of 1 mg/ml in a solution containing PBS (pH 7.4), 10% glycerol, and 0.02% sodium azide.
Storage Procedures
For short-term storage (up to 1 month), store at 4°C. For extended storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This antibody has been validated for ELISA and Western blot analysis, demonstrating its specificity and reactivity. However, optimal working dilutions should be determined empirically for each application. A starting dilution of 1:1000 is recommended.
Synonyms
Protein phosphatase 1F, Ca(2+)/calmodulin-dependent protein kinase phosphatase, CaM-kinase phosphatase, CaMKPase, Partner of PIX 2, Protein fem-2 homolog, hFem-2, KIAA0015, POPX2, PPM1F, CAMKP, FEM-2.
Purification Method
PPM1F antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT6D11AT.
Immunogen
Anti-human PPM1F mAb, clone PAT6D11AT, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human PPM1F protein 1-454 amino acids purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and k light chain.

Q&A

What is PPM1F and what are its primary biological functions?

PPM1F is a serine/threonine phosphatase belonging to the PP2C family of protein phosphatases that dephosphorylates threonine motifs in various cellular contexts . This 50 kDa protein (453 amino acids) is ubiquitously expressed in various tissues and organs throughout the body . Functionally, PPM1F serves as a critical regulator in multiple cellular processes including:

  • Integrin activity regulation through control of the T788/T789 phospho-switch in the integrin β1 cytoplasmic tail

  • Neuronal excitability modulation in the medial prefrontal cortex with implications for depression-related behaviors

  • AMPK signaling pathway regulation via dephosphorylation events

  • Potential roles in cancer progression, though with context-dependent effects across different cancer types

The diverse functions of PPM1F make it a compelling target for research in cell adhesion, neuropsychiatry, and oncology fields.

Which experimental models are most appropriate for studying PPM1F functions?

Based on the available research data, several experimental models have proven effective for studying PPM1F function:

Cellular Models:

  • Normal human dermal fibroblasts (NHDF) for adhesion and integrin studies

  • Human glioblastoma A172 cells for integrin activity assays

  • HEK-293 cells for basic expression experiments

  • Gastric cancer cell lines (AGS and MKN-28) for cancer-related studies

  • U2OS, A375, HepG2, Jurkat, and K-562 cells have all demonstrated detectable PPM1F expression in Western blot applications

Animal Models:

  • Mouse models with targeted manipulation of PPM1F expression in specific brain regions (e.g., medial prefrontal cortex) for neuropsychiatric research

  • Chronic unpredictable stress (CUS) mouse models to study PPM1F involvement in depression

The selection of an appropriate model should be guided by the specific aspect of PPM1F biology under investigation.

What is the recommended protocol for Western blot detection of PPM1F?

For optimal Western blot detection of PPM1F, researchers should follow these methodological guidelines:

  • Sample preparation: Extract proteins using standard lysis buffer from cultured cells or tissue samples

  • Antibody selection: Use validated anti-PPM1F antibodies such as monoclonal antibody 68626-1-Ig for human samples

  • Antibody dilution: Apply recommended dilution factors (1:5000-1:50000) for Western blot applications, though titration in each specific system is advised

  • Expected molecular weight: Look for bands at approximately 50 kDa, which corresponds to the observed molecular weight of PPM1F

  • Controls: Include positive control samples such as U2OS, A375, HEK-293, HepG2, Jurkat, or K-562 cells, which all show reliable PPM1F expression

For validation studies, researchers should note that PPM1F detection has been successful with several antibodies, including rabbit polyclonal antibodies (e.g., ab156222) at 1:1000 dilution with appropriate secondary antibodies .

How can PPM1F activity be modulated for functional studies?

To effectively manipulate PPM1F activity for functional studies, researchers have employed several methodological approaches:

Genetic Approaches:

  • RNA interference: Short hairpin RNA (shRNA) for PPM1F knockdown in neural tissue has demonstrated effective modulation of PPM1F expression with consequent behavioral effects

  • CRISPR/Cas9 gene editing: Complete deletion of PPM1F gene in A172 cells showed pronounced effects on integrin activity and cell adhesion

  • Viral vector-mediated overexpression: Adeno-associated virus strategies for PPM1F overexpression in specific neuronal populations

Target Cell Types for Specific Research Questions:

  • Excitatory neurons in the medial prefrontal cortex for depression-related research

  • Fibroblasts and glioblastoma cells for integrin and adhesion studies

  • Gastric cancer cells for investigating miRNA regulation of PPM1F

Importantly, while modulating PPM1F expression, researchers should verify that observed phenotypes are not due to altered expression of related proteins (e.g., integrin subunits or cytosolic focal adhesion proteins) .

What downstream pathways and molecular partners should be assessed in PPM1F research?

When investigating PPM1F functions, several critical downstream pathways and interaction partners should be evaluated:

Integrin Signaling Pathway:

  • Active integrin β1 levels using specific antibodies against the active conformation

  • Talin recruitment to focal adhesions

  • FilaminA interactions, which functionally connect to PPM1F effects

Neuropsychiatric Molecular Mechanisms:

  • CREB-binding protein (CBP)/E1A-associated protein (p300) expression

  • AMPK phosphorylation status

  • Neuronal excitability measurements via electrophysiological recordings

  • Proinflammatory cytokine expression as a secondary effect

Cancer-Related Pathways:

  • miR-590 expression levels, which negatively regulate PPM1F in gastric cancer

  • Assessment of 3'UTR interactions using luciferase reporter systems

The comprehensive assessment of these molecular partners provides a more complete understanding of PPM1F's functional impact in specific biological contexts.

How can phosphatase activity of PPM1F be directly measured?

For direct measurement of PPM1F phosphatase activity, researchers should consider these methodological approaches:

  • In vitro phosphatase assays using recombinant PPM1F protein and phosphorylated substrate peptides (particularly those containing the T788/T789 motif from integrin β1)

  • Cellular phosphorylation status assessment by:

    • Phospho-specific antibodies against known PPM1F substrates

    • Western blotting for phosphorylated AMPK, which shows hyperphosphorylation upon PPM1F knockdown

    • Monitoring the phosphorylation status of the T788/T789 sites on integrin β1

  • Functional readouts of phosphatase activity:

    • Cell adhesion assays on integrin ligands, which increase with PPM1F depletion

    • Measurement of active integrin β1 using conformation-specific antibodies

    • Assessment of neuronal excitability in response to PPM1F modulation

Researchers should be aware that Mg2+/Mn2+ dependence is a characteristic feature of PPM1F activity, and buffer conditions should be optimized accordingly.

How do PPM1F expression patterns differ between normal and pathological states?

PPM1F expression exhibits notable differences between normal and pathological states, with some intriguing tissue-specific patterns:

Neuropsychiatric Disorders:

  • Significant decreases in PPM1F expression in the medial prefrontal cortex of mice exposed to chronic unpredictable stress, a model of depression

  • Contrastingly, previous research found increased PPM1F expression in the hippocampus associated with depression and anxiety

Cancer Biology:

  • Contradictory patterns observed across different cancer types:

    • Upregulation reported in hepatocellular carcinoma (HCC) and breast cancer

    • Downregulation observed in gastric cancer compared to adjacent normal tissues (53.33% vs 86.67% positive rate)

    • High expression positively associated with poor survival and tumor recurrence in some cancer patients

These contradictory findings suggest that PPM1F expression and function are highly context-dependent and may vary significantly between different tissues, pathological conditions, and even specific brain regions within the same disorder.

What are the common challenges in interpreting PPM1F knockout or knockdown experiments?

Interpreting PPM1F knockout or knockdown experiments presents several methodological challenges that researchers should address:

  • Distinguishing primary from secondary effects:

    • PPM1F knockdown affects integrin activity without altering expression of integrin subunits or focal adhesion proteins

    • In neural tissue, PPM1F knockdown leads to microglial activation and upregulation of proinflammatory cytokines as secondary effects

  • Phenotypic paradoxes:

    • PPM1F knockout cells show enhanced adhesion but compromised cell spreading, suggesting complex effects on cell-matrix interactions

    • PPM1F knockdown reduces neuronal excitability but increases depression-related behaviors, requiring careful mechanistic dissection

  • Tissue-specific effects:

    • Opposite expression patterns in hippocampus versus medial prefrontal cortex in depression models highlight the need for region-specific analyses

    • Cancer-type specific expression patterns demand careful selection of appropriate control tissues

To address these challenges, researchers should employ comprehensive approaches including:

  • Multiple readouts of cellular function

  • Time-course experiments to distinguish primary from secondary effects

  • Rescue experiments to confirm specificity of observed phenotypes

  • Careful selection of appropriate control cells or tissues

What is the relationship between PPM1F and microRNA regulation in research contexts?

The relationship between PPM1F and microRNA regulation represents an important regulatory mechanism with significant research implications:

Established microRNA regulators of PPM1F:

  • miR-590-3p shows strong negative correlation with PPM1F expression in gastric cancer samples and cell lines

  • miR-186-5p, miR-200b, and miR-429 also demonstrate negative correlation with PPM1F expression in gastric cancer

Experimental validation approaches:

  • Spearman correlation analysis between miRNA and PPM1F expression in clinical samples

  • Transfection of miRNA mimics or inhibitors to modulate PPM1F expression levels

  • Luciferase reporter assays with wild-type and mutant 3'UTR constructs of PPM1F to validate direct binding

Research applications:

  • Understanding post-transcriptional regulation of PPM1F when genetic alterations or methylation changes cannot explain expression differences

  • Exploring therapeutic targeting of miRNA-PPM1F interactions in relevant disease contexts

  • Explaining tissue-specific or context-dependent regulation of PPM1F

This microRNA regulatory mechanism is particularly important when explaining PPM1F downregulation in contexts where genetic alterations (amplification, deletion, mutation) or epigenetic modifications (methylation) do not account for the observed expression changes .

How does PPM1F research connect to integrin biology and cell adhesion studies?

PPM1F has emerged as a key regulator of integrin biology and cell adhesion through specific molecular mechanisms:

Core molecular mechanism:

  • PPM1F controls the T788/T789 phospho-switch in the integrin β1 cytoplasmic tail

  • This phosphorylation state directly affects integrin activation status and downstream signaling

Experimental evidence and phenotypes:

  • PPM1F knockdown in multiple cell types (293T, NHDF, A172) consistently enhances cell adhesion to integrin ligands

  • PPM1F-depleted cells show elevated levels of active integrin β1

  • Characteristic "active integrin belt" at cell periphery observed in PPM1F knockout cells

  • Enhanced talin recruitment to focal adhesions occurs with PPM1F depletion

Research implications:

  • PPM1F represents a novel target for modulating integrin activity in diverse contexts

  • The PPM1F-integrin axis provides insight into basic mechanisms of cell-matrix interactions

  • Compromised cell spreading despite enhanced adhesion in PPM1F-depleted cells suggests complex effects on the dynamic regulation of cell-matrix interactions

This connection to integrin biology opens research opportunities in wound healing, tissue engineering, and metastasis studies where cell adhesion plays critical roles.

What is the evidence linking PPM1F dysfunction to neuropsychiatric disorders?

The connection between PPM1F dysfunction and neuropsychiatric disorders is supported by several lines of evidence:

Genetic associations:

  • Six PPM1F single-nucleotide polymorphisms affect the association between PTSD symptom severity and cortical thickness in frontal brain regions

  • These genetic variants may influence neural integrity of the prefrontal cortex

Expression changes in depression models:

  • Significant decrease in PPM1F expression in the medial prefrontal cortex of mice exposed to chronic unpredictable stress

  • Previous research found increased PPM1F expression in the hippocampus associated with depression and anxiety

Functional validation:

  • shRNA-mediated knockdown of PPM1F in the mPFC produces depression-related behaviors

  • Overexpression of PPM1F produces antidepressant effects and ameliorates stress responses

  • PPM1F knockdown decreases excitability of pyramidal neurons in the mPFC

  • Restoring neuronal excitability reverses depression-related behaviors induced by PPM1F knockdown

Molecular mechanisms:

  • PPM1F knockdown reduces expression of CBP/p300 histone acetyltransferase

  • PPM1F depletion induces AMPK hyperphosphorylation

  • These molecular changes lead to microglial activation and increased proinflammatory cytokines

These findings establish PPM1F as a potential therapeutic target for depression and related disorders, with particular emphasis on its region-specific effects in the brain.

How can PPM1F research contribute to cancer biology?

PPM1F research has revealed complex and sometimes contradictory roles in cancer biology that warrant further investigation:

Expression patterns across cancer types:

  • Upregulation reported in hepatocellular carcinoma and breast cancer

  • Downregulation observed in gastric cancer compared to adjacent normal tissues

  • High expression positively associated with poor survival and tumor recurrence in some cancer patients

Regulatory mechanisms in cancer:

  • In gastric cancer, PPM1F is negatively regulated by miR-590, which shows increased expression in tumor samples

  • The 3'UTR of PPM1F is targeted by multiple miRNAs (miR-590-3p, miR-186-5p, miR-200b, miR-429)

  • No significant alterations in PPM1F at genetic or methylation levels were found in gastric cancer

Potential as biomarker:

  • PPM1F might serve as a potential biomarker in hepatocellular carcinoma patients

  • Loss of PPM1F expression predicts tumor recurrence in some cancers

Research implications:

  • The context-dependent expression patterns suggest tissue-specific roles of PPM1F in cancer development

  • The connection to integrin signaling may link PPM1F to cancer cell adhesion, migration, and invasion

  • The miRNA regulatory network offers potential therapeutic targets

These contradictory findings highlight the need for cancer-type specific investigations of PPM1F function and careful consideration of tissue context when designing cancer-related studies.

What are common technical challenges when working with PPM1F antibodies?

Researchers working with PPM1F antibodies should be aware of several technical challenges:

Antibody specificity concerns:

  • Validation through PPM1F knockdown/knockout controls is essential for confirming antibody specificity

  • Multiple antibodies with different host species are available (mouse monoclonal, rabbit polyclonal) and may perform differently across applications

Optimal working conditions:

  • Wide recommended dilution range (1:5000-1:50000) for Western blot applications suggests variable sensitivity across systems

  • Titration in each testing system is recommended to obtain optimal results

  • Sample-dependent performance requires validation with positive controls

Detection strategies:

  • For Western blot applications, positive detection has been validated in multiple cell lines (U2OS, A375, HEK-293, HepG2, Jurkat, K-562, human peripheral blood platelets)

  • For IHC applications in tissues, appropriate antigen retrieval methods should be optimized

Storage and handling:

  • Store antibodies at -20°C for stability

  • For the 68626-1-Ig antibody, aliquoting is unnecessary for -20°C storage

  • Some preparations contain 0.1% BSA which may affect certain applications

Researchers are advised to include appropriate positive controls and validation experiments when using PPM1F antibodies in new experimental systems.

What research gaps remain in understanding PPM1F function across different tissues?

Despite significant advances, several important research gaps remain in our understanding of PPM1F biology:

Tissue-specific regulation:

  • The mechanisms underlying opposite expression patterns of PPM1F in different brain regions during depression remain unclear

  • The factors determining cancer-type specific expression patterns (up vs. down) need further investigation

Substrate specificity:

  • Beyond integrin β1 T788/T789, the complete substrate spectrum of PPM1F across tissues is not fully characterized

  • How substrate preferences might differ between tissues remains largely unexplored

Regulatory network:

  • While miRNA regulation is established in gastric cancer, the upstream regulators of PPM1F in other tissues are not well defined

  • The complete transcriptional regulatory network controlling PPM1F expression is incompletely understood

Therapeutic potential:

  • Methods for selective pharmacological modulation of PPM1F activity (rather than expression) need development

  • Tissue-specific targeting strategies to avoid unintended effects across systems

Translational relevance:

  • The clinical significance of PPM1F alterations in neuropsychiatric disorders requires human validation studies

  • The prognostic value of PPM1F in different cancer types needs systematic evaluation

Addressing these gaps represents important directions for future PPM1F research.

What emerging technologies might enhance PPM1F research in the near future?

Several emerging technologies hold promise for advancing PPM1F research:

Advanced genetic manipulation techniques:

  • CRISPR activation/inhibition systems for endogenous PPM1F modulation without complete knockout

  • Cell-type specific and inducible Cre-lox systems for temporal control of PPM1F expression in animal models

  • Base editing for introducing specific PPM1F mutations to study structure-function relationships

Spatial resolution technologies:

  • Spatial transcriptomics to map PPM1F expression patterns at high resolution within tissues

  • Super-resolution microscopy for detailed visualization of PPM1F interactions at focal adhesions

  • Proximity labeling methods (BioID, APEX) to identify tissue-specific PPM1F interactors

Functional approaches:

  • Phosphoproteomic analysis to comprehensively identify PPM1F substrates across tissues

  • Single-cell technologies to understand cell-type specific functions

  • Organoid models to study PPM1F in more physiologically relevant 3D systems

Translational methods:

  • Development of small molecule modulators of PPM1F activity

  • Patient-derived xenografts to study PPM1F in personalized cancer models

  • Integration of PPM1F status with multi-omics datasets for predictive biomarker development

These technological advances will likely contribute to a more nuanced understanding of PPM1F biology and potentially reveal new therapeutic opportunities targeting this phosphatase in various disease contexts.

Product Science Overview

Gene and Protein Information
  • Gene Location: In humans, the PPM1F gene is located on chromosome 22 (22q11.22), while in mice, it is found on chromosome 16 .
  • Protein Function: PPM1F dephosphorylates and deactivates CaM-kinase II, IV, and I, which are activated upon phosphorylation by CaM-kinase kinase. This dephosphorylation process is crucial for regulating various cellular functions .
Biological Role and Mechanism

PPM1F interacts with Rho guanine nucleotide exchange factors (PIX), thereby blocking the effects of p21-activated kinase 1 (PAK), a protein kinase that mediates biological effects downstream of Rho GTPases . One of the substrates of PPM1F is Calcium/calmodulin-dependent protein kinase II gamma (CAMK2G/CAMK-II), which plays a role in various cellular processes, including apoptosis .

Clinical Significance

Overexpression of PPM1F or CAMK2G has been shown to mediate caspase-dependent apoptosis, highlighting its potential role in programmed cell death . Additionally, PPM1F is associated with diseases such as cholestasis and benign recurrent intrahepatic cholestasis .

Research and Applications

The mouse anti-human PPM1F antibody is used in various research applications to study the enzyme’s function and its role in different cellular pathways. This antibody helps in identifying and quantifying PPM1F in human tissues, aiding in the understanding of its biological significance and potential therapeutic targets .

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.