TMEM88 Antibody

Shipped with Ice Packs
In Stock

Description

Product Overview

The TMEM88 antibody is a polyclonal or monoclonal antibody designed to detect the transmembrane protein 88 (TMEM88), a tumor suppressor implicated in various cancers. Key specifications include:

ParameterDetails
SKUA12951 (Boster Bio)
Reactive SpeciesHuman, Mouse, Rat
HostRabbit
ApplicationsELISA, Immunofluorescence (IF), Immunohistochemistry (IHC-P), Western Blot (WB)
StorageStable at 4°C for 3 months; -20°C for up to 1 year
Concentration1 μg/mL (WB), 2.5 μg/mL (IHC-P), 20 μg/mL (IF)

Validation and Applications

The TMEM88 antibody has been rigorously validated across multiple platforms:

Validation Methods

AssayConditionsResults
Western BlotHuman brain tissue lysate, 1 μg/mLDetects TMEM88 protein; blocked by specific peptides
ImmunohistochemistryMouse brain tissue, 2.5 μg/mLStains TMEM88 in tissue sections; validated for localization studies
ImmunofluorescenceMouse brain tissue, 20 μg/mLVisualizes TMEM88 subcellular distribution (e.g., cytoplasmic vs. nuclear)

Research Findings and Clinical Relevance

The TMEM88 antibody has enabled critical insights into TMEM88’s role in cancer biology and therapeutic resistance:

Mechanistic Insights

  • Wnt/β-Catenin Signaling: TMEM88 inhibits Wnt signaling by interacting with Dishevelled-1 (Dvl-1), reducing β-catenin activity. Antibody-based studies confirm its role in suppressing oncogenic pathways in thyroid and ovarian cancers .

  • Subcellular Localization:

    • Cytosolic TMEM88: Promotes invasion/metastasis in NSCLC .

    • Nuclear TMEM88: Associated with better outcomes in non-triple-negative breast cancer .

Technical Considerations and Limitations

  • Cross-Reactivity: Validated for human, mouse, and rat; other species require confirmation .

  • Assay Optimization: Dilutions vary by application; e.g., 1 μg/mL for WB vs. 20 μg/mL for IF .

  • Clinical Translation: While TMEM88 is a prognostic biomarker, the antibody’s utility in routine diagnostics remains under investigation .

Future Directions

  • Therapeutic Targeting: Antibody-based studies may guide TMEM88 modulation strategies to enhance chemotherapy sensitivity (e.g., in ovarian cancer) .

  • Multicancer Biomarker: Further validation across tumor types to establish TMEM88 as a pan-cancer prognostic marker .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. The delivery time may vary depending on the method of purchase or location. For specific delivery times, please consult your local distributors.
Synonyms
TMEM88; TMEM88A; Transmembrane protein 88
Target Names
TMEM88
Uniprot No.

Target Background

Function
TMEM88 Antibody inhibits the Wnt/beta-catenin signaling pathway, which plays a crucial role in heart development. It functions downstream of GATA factors in the pre-cardiac mesoderm, specifying lineage commitment for cardiomyocyte development.
Gene References Into Functions
  1. TMEM88 inhibits TGF-beta1-stimulated cell proliferation, migration, and extracellular matrix expression in keloid fibroblasts. PMID: 28946191
  2. TMEM88, CCL14, and CLEC3B genes exhibit stability and are valuable in predicting the survival and palindromia time of hepatocellular carcinoma. These genes serve as potential prognostic markers, contributing to improved patient outcomes and survival. PMID: 28718365
  3. Research indicates that platinum resistance in ovarian cancer is associated with TMEM88 overexpression, regulated through decreased promoter methylation. These findings suggest that TMEM88 acts as an inhibitor of Wnt signaling, contributing to the development of platinum resistance. PMID: 27374141
  4. TMEM88 promotes triple-negative breast cancer cell invasion by interacting with DVL1. PMID: 26325443
  5. Mislocalization of TMEM88 to the cytosol in non-small cell lung cancer cells eliminates its Wnt pathway regulatory properties, leading to increased invasion and metastasis by activating the p38-GSK3beta-Snail signaling pathway. PMID: 26359454
  6. TMEM88 is critical for heart development, functioning downstream of GATA factors in the pre-cardiac mesoderm to specify lineage commitment of cardiomyocytes. PMID: 23924634
  7. miRNA-708 acts as an oncogene, contributing to tumor growth and disease progression by directly downregulating TMEM88. PMID: 22573352
  8. TMEM88 associates with Dvl proteins and regulates Wnt signaling in a context-dependent manner. PMID: 21044957

Show More

Hide All

Database Links

HGNC: 32371

OMIM: 617813

KEGG: hsa:92162

STRING: 9606.ENSP00000301599

UniGene: Hs.389669

Protein Families
TMEM88 family
Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What is TMEM88 and why is it significant in cellular signaling research?

TMEM88 is a two-transmembrane-type protein that plays a critical role in regulating the Wnt signaling pathway. Its significance derives from its C-terminal tail that binds to the PDZ domain of Dishevelled (Dvl), a key component in Wnt signaling pathways . TMEM88 attenuates Wnt/β-catenin signaling induced by Wnt-1 ligand in a dose-dependent manner, while knockdown of TMEM88 by RNAi increases Wnt activity . This regulatory function makes TMEM88 an important research target in developmental biology, cancer research, and metabolic disease studies.

The human canonical protein consists of 159 amino acid residues with a molecular mass of 17.3 kDa . At least two isoforms of TMEM88 are known to exist, with most antibodies recognizing both isoforms .

What is the subcellular localization pattern of TMEM88?

TMEM88 exhibits complex subcellular localization patterns that are critical to understand when designing immunostaining experiments:

  • Plasma membrane: TMEM88 is prominently found at the cell surface, where it forms membrane-associated puncta with dynamic movements and internalization

  • Golgi apparatus: Significant expression in Golgi membranes has been consistently observed

  • Multivesicular bodies (MVBs): TMEM88 localizes to perinuclear MVBs that can be visualized as EEA1-positive structures resistant to digitonin permeabilization

  • Endocytic vesicles: Present in trafficking vesicles during internalization

This multi-compartment localization pattern suggests that TMEM88 trafficking between cellular compartments may be integral to its function. Importantly, nuclear versus cytoplasmic localization appears to correlate with different disease outcomes, particularly in breast cancer where cytoplasmic TMEM88 promotes tumor progression while nuclear TMEM88 may suppress tumors .

What applications are TMEM88 antibodies commonly used for?

TMEM88 antibodies are utilized across multiple experimental applications:

ApplicationCommon Use CasesTechnical Considerations
Western BlotProtein expression quantificationObserved MW: ~68 kDa (despite calculated 17.3 kDa)
ImmunohistochemistryTissue localization studiesOften requires antigen retrieval optimization
ImmunofluorescenceSubcellular localizationDigitonin permeabilization reveals MVB-specific staining
ELISAQuantitative protein detectionMost common application for commercial antibodies
Flow CytometryCell population analysisUsed for apoptosis studies in AML-12 cells

While the calculated molecular weight of TMEM88 is 17.3 kDa, the observed molecular weight in Western blots is typically around 68 kDa , which may reflect post-translational modifications or protein complexes.

How should sample preparation be optimized for detecting TMEM88 in different subcellular compartments?

Sample preparation is critical for accurate detection of TMEM88 across its various subcellular localizations:

For MVB-specific detection:

  • Use digitonin permeabilization (before fixation) to remove cytosolic proteins while preserving membrane-associated TMEM88

  • Co-stain with the MVB marker EEA1, focusing on digitonin-resistant EEA1+ structures as a proxy for MVBs

  • Western blot analysis of digitonin-resistant versus soluble fractions can confirm MVB-associated TMEM88 (it co-segregates with membrane markers like NaK-ATPase)

For plasma membrane visualization:

  • Live imaging using total internal reflection fluorescence microscopy reveals membrane-associated puncta with dynamic movements

  • N-terminal GFP fusion constructs (GFP-TMEM88) are effective for tracking membrane localization and internalization dynamics

For complete subcellular profiling:

  • Immunoelectron microscopy using gold-conjugated anti-TMEM88 antibodies provides high-resolution localization data

  • Subcellular fractionation followed by Western blotting of distinct fractions helps quantify distribution patterns

These methodological approaches allow researchers to specifically track TMEM88 across different cellular compartments, which is essential for understanding its context-dependent functions.

What are the key considerations for selecting appropriate TMEM88 antibodies for specific applications?

When selecting TMEM88 antibodies, researchers should consider:

  • Epitope location: Antibodies generated against C-terminal epitopes (amino acids 110-160) are particularly effective for studying TMEM88 function, as this region contains the PDZ-binding domain critical for Wnt signaling regulation

  • Clonality considerations:

    • Polyclonal antibodies offer broader epitope recognition but may have batch-to-batch variation

    • Monoclonal antibodies provide consistent results but may miss certain conformational states

  • Isoform recognition: Verify whether the antibody recognizes all known TMEM88 isoforms if your research requires comprehensive detection

  • Application-specific validation:

    • For Western blot: Validate using positive controls (human brain tissue lysate shows reliable detection)

    • For IHC/IF: Test fixation conditions as they significantly affect epitope accessibility

    • For live cell imaging: Consider non-perturbing antibody fragments or fluorescent protein fusions

  • Use of blocking peptides: When testing antibody specificity, include controls using the immunizing peptide to confirm binding specificity

What methodological approaches can resolve the discrepancy between calculated (17.3 kDa) and observed (~68 kDa) molecular weights of TMEM88?

The significant difference between calculated (17.3 kDa) and observed (approximately 68 kDa) molecular weights of TMEM88 in Western blots presents a methodological challenge . To address this discrepancy, researchers should consider:

  • Denaturation optimization:

    • Test different denaturation conditions (varying SDS concentrations, reducing agents, and heating temperatures)

    • Compare boiled versus non-boiled samples to detect potential heat-induced aggregation

  • Post-translational modification analysis:

    • Use phosphatase or glycosidase treatments prior to Western blotting to identify potential modifications

    • Apply mass spectrometry to characterize post-translational modifications that may affect migration

  • Cross-linking studies:

    • Use chemical cross-linking followed by Western blotting to determine if TMEM88 exists in stable complexes

    • Perform blue native PAGE to preserve protein complexes and compare with denaturing conditions

  • Alternative detection methods:

    • Combine immunoprecipitation with mass spectrometry to confirm protein identity

    • Use epitope-tagged expression constructs (such as GFP-TMEM88) as controls

  • Validation with knockout/knockdown controls:

    • Include TMEM88 siRNA-treated samples as negative controls to confirm band specificity

    • Use tissue from knockout models when available as definitive controls

These methodological approaches can help distinguish between true TMEM88 signal and potential artifacts or complex formation.

How does TMEM88 mechanistically regulate the Wnt/β-catenin signaling pathway?

TMEM88 regulates Wnt/β-catenin signaling through direct interaction with Dishevelled (Dvl) proteins via a specific C-terminal mechanism:

  • PDZ domain interaction:

    • The C-terminal tripeptide Val-Trp-Val (VWV) sequence of TMEM88 directly binds to the PDZ domain of Dvl

    • This interaction can be quantitatively measured using fluorescence spectroscopy, where Trp fluorescent polarization of the Val-Trp-Val tripeptide shows measurable changes upon Dvl-1 PDZ domain binding

  • Signaling inhibition mechanism:

    • TMEM88 binding promotes Wnt signalosome degradation in multivesicular bodies (MVBs)

    • In experimental systems, TMEM88 inhibits Siamois promoter-driven luciferase activity induced by Xdsh (Xenopus Dishevelled) but not by β-catenin, indicating it acts at the level of Dishevelled rather than downstream components

  • Context-dependent regulation:

    • In Xenopus secondary axis assays, co-injection of Xdsh and TMEM88 mRNAs abolishes the formation of complete secondary axes induced by Xdsh alone

    • Deletion of the C-terminal PDZ-binding motif (TMEM88-ΔC) eliminates the inhibitory effect on Wnt signaling

  • Functional validation approaches:

    • Luciferase reporter assays using TCF/LEF or Siamois promoter-driven constructs provide quantitative measurement of pathway inhibition

    • GFP-tagged Dvl localization studies can visualize how TMEM88 affects Dvl distribution patterns

These mechanistic insights explain how TMEM88 functions as a context-dependent regulator of Wnt signaling, rather than as a simple on/off switch.

How does TMEM88 function differ across various cancer types, and what methodologies best capture these differences?

TMEM88 exhibits striking context-dependent functions across different cancer types:

Cancer TypeTMEM88 ExpressionFunctional RoleMethodological Approaches
Ovarian CancerIncreased in platinum-resistant tumorsPromotes chemoresistance and cell dormancysiRNA-mediated knockdown restores platinum sensitivity
Breast CancerContext-dependentCytoplasmic: promotes tumors
Nuclear: suppresses tumors
Subcellular fractionation and compartment-specific analysis
Lung Cancer (NSCLC)Variable expressionGenerally tumor-suppressiveMethylation analysis shows higher TMEM88 methylation in NSCLC (82.2% ± 10.3%) vs. normal tissue (65.9% ± 7.2%)
Thyroid CancerSignificantly reducedTumor-suppressiveRestoration via vector transfection suppresses proliferation and invasion
Bladder CancerVariable expressionTumor-suppressiveOverexpression inhibits bladder cancer growth in xenograft models

To properly study these context-dependent functions, researchers should employ:

  • Subcellular localization analysis:

    • Distinguish between nuclear and cytoplasmic fractions, particularly in breast cancer where localization correlates with different outcomes

    • Use confocal microscopy with co-staining for compartment markers

  • Methylation analysis:

    • Quantify TMEM88 promoter methylation using bisulfite sequencing or methylation-specific PCR

    • Treat cells with demethylating agents like 5-aza-2'-deoxycytidine (DAC) to evaluate functional changes

  • Pathway integration analysis:

    • Analyze correlation between TMEM88 and Wnt pathway components (c-Myc, β-catenin) using TCGA database mining

    • Perform rescue experiments by modulating both TMEM88 and downstream effectors

  • Chemoresistance models:

    • Develop platinum-resistant cell lines to study TMEM88's role in drug resistance

    • Use patient-derived xenografts from treatment-resistant tumors

These methodological approaches help capture the complex and sometimes contradictory roles of TMEM88 across different cancer contexts.

What role does TMEM88 play in lipid metabolism and how can this be experimentally investigated?

TMEM88 exerts significant regulatory effects on lipid metabolism, particularly relevant in the context of Non-Alcoholic Fatty Liver Disease (NAFLD):

  • Regulation of key lipid metabolism factors:

    • TMEM88 upregulates PPAR-α and its downstream target ACOX-1 (promoting fatty acid oxidation)

    • TMEM88 downregulates SREBP-1c and its downstream target FASN (reducing fatty acid synthesis)

    • This dual regulation suggests TMEM88 shifts cellular metabolism away from lipid accumulation

  • Experimental approaches to study this function:

    a) In vitro models:

    • Free Fatty Acid (FFA)-induced AML-12 hepatocyte cells transfected with pEGFP-C1-TMEM88 or TMEM88 siRNA

    • Quantification of lipid synthesis and metabolism markers using RT-qPCR and Western blotting

    b) In vivo models:

    • Methionine and Choline-Deficient (MCD) diet-induced NAFLD mouse models

    • H&E staining and immunohistochemistry of liver tissue to assess steatosis

    • Analysis of lipid metabolism markers in tissue samples

  • Functional consequences:

    • TMEM88 accelerates the apoptotic rate of FFA-induced AML-12 cells (measurable via Annexin V-FITC/PI double staining flow cytometry)

    • TMEM88 inhibits proliferation of FFA-stimulated hepatocytes (detectable via EdU staining)

    • These effects may contribute to limiting the expansion of steatotic liver cells

  • Signaling pathway integration:

    • TMEM88 mediates these effects through inhibition of the Wnt/β-catenin signaling pathway

    • Experimental validation can be performed using TCF/LEF reporter assays in the presence of metabolic stressors

These findings suggest TMEM88 may represent a novel therapeutic target for metabolic disorders involving dysregulated lipid metabolism.

What are the most effective approaches for studying TMEM88 trafficking dynamics in living cells?

Investigating the dynamic trafficking of TMEM88 between cellular compartments requires specialized approaches:

  • Fluorescent protein fusion constructs:

    • N-terminal GFP fusion (GFP-TMEM88) effectively visualizes plasma membrane localization and internalization

    • Compare with C-terminal tags to ensure functionality is preserved

    • Validate that fusion proteins maintain normal interactions with binding partners (e.g., Dvl)

  • Live cell imaging techniques:

    • Total Internal Reflection Fluorescence (TIRF) microscopy reveals membrane-associated puncta with dynamic movements and internalization events

    • Spinning disk confocal microscopy allows longer-term imaging with reduced phototoxicity

    • Dual-color imaging with markers for different compartments (e.g., Rab proteins) enables tracking of TMEM88 through the endosomal system

  • Photoactivatable/photoconvertible tags:

    • Fusion with photoconvertible fluorescent proteins (e.g., mEos, Dendra2) enables pulse-chase visualization of specific protein populations

    • This approach can determine the half-life of TMEM88 in different compartments and trafficking rates

  • Cargo trafficking assays:

    • Co-tracking with known endocytic cargo (transferrin, EGF) to determine if TMEM88 follows canonical trafficking routes

    • Pharmacological inhibitors of different trafficking pathways can identify the mechanisms of TMEM88 movement

  • FRAP (Fluorescence Recovery After Photobleaching) analysis:

    • Measure the mobility and exchange rates of TMEM88 in different compartments

    • Compare recovery kinetics between normal and disease conditions

These approaches provide complementary information about the complex trafficking patterns of TMEM88, which may be critical to its function in different cellular contexts.

How can researchers resolve contradictory findings about TMEM88's role in different experimental systems?

The literature contains seemingly contradictory findings regarding TMEM88's function across different experimental systems. To address these contradictions, researchers should:

  • Systematically analyze context-dependent variables:

    VariableMethodological ApproachExample from Literature
    Cell/tissue typeDirect comparison in multiple cell linesTMEM88 is tumor-suppressive in thyroid cancer but promotes chemoresistance in ovarian cancer
    Subcellular localizationFraction-specific analysisCytoplasmic vs. nuclear TMEM88 have opposite correlations with lymph node metastasis in breast cancer
    Isoform expressionIsoform-specific detection methodsDifferent TMEM88 isoforms may have distinct functions
    Wnt pathway activation stateBaseline pathway analysis before manipulationEffects may differ between Wnt-active vs. Wnt-inactive backgrounds
  • Develop standardized reporting:

    • Document complete experimental conditions including cell density, passage number, and media composition

    • Report antibody catalog numbers, dilutions, and validation methods

    • Specify exact genetic constructs used (full sequences preferred)

  • Combinatorial approaches:

    • Simultaneously manipulate TMEM88 and potential modifying factors

    • Use rescue experiments with wild-type vs. mutant constructs lacking specific domains

    • Employ dose-response studies rather than single-concentration experiments

  • Systems biology perspective:

    • Analyze TMEM88 in the context of broader signaling networks using phosphoproteomics

    • Apply computational modeling to predict context-dependent outcomes

    • Use correlation analysis with gene expression databases to identify potential modifiers

  • Cross-validation between in vitro and in vivo systems:

    • Confirm cell line findings in primary cells and tissue samples

    • Develop tissue-specific conditional knockout models to address developmental compensation

These approaches can help resolve contradictory findings by identifying the specific conditions under which TMEM88 exerts different functions.

What methodological approaches can advance our understanding of TMEM88's role in multivesicular body (MVB) biology?

TMEM88's presence in multivesicular bodies (MVBs) suggests an important role in protein trafficking and degradation that requires specialized methodological approaches:

  • Advanced imaging techniques:

    • Super-resolution microscopy (STORM, PALM) to visualize TMEM88 distribution within MVB subdomains

    • Correlative light and electron microscopy (CLEM) to connect fluorescent signals with ultrastructural features

    • Live-cell imaging of GFP-TMEM88 with markers for MVB formation and maturation

  • Biochemical isolation of MVBs:

    • Density gradient fractionation to isolate MVB-enriched fractions

    • Immunomagnetic isolation using antibodies against MVB markers

    • Proteomic analysis of isolated MVBs to identify TMEM88-interacting proteins

  • Functional perturbation of MVB machinery:

    • ESCRT complex component knockdown/knockout to determine dependence of TMEM88 trafficking on canonical MVB formation

    • Manipulation of Rab proteins (particularly Rab7) to assess effects on TMEM88 localization

    • Lysosomal inhibition to distinguish between degradative and recycling MVB populations

  • Analysis of exosomal TMEM88:

    • Isolation of extracellular vesicles using ultracentrifugation or size-exclusion chromatography

    • Western blot and proteomic analysis of exosomal fractions for TMEM88 content

    • Functional studies of TMEM88-containing exosomes in recipient cells

  • MVB cargo sorting studies:

    • Investigation of TMEM88's potential role in sorting other proteins to MVBs

    • Analysis of ubiquitination patterns associated with TMEM88 trafficking

    • CRISPR-Cas9 screening for genes affecting TMEM88 MVB localization

These methodologies can help elucidate TMEM88's role in MVB biology, potentially revealing new functions in protein degradation, signaling regulation, and intercellular communication.

What are the most promising approaches for developing TMEM88 as a therapeutic target in cancer?

Based on current research, several approaches show promise for targeting TMEM88 therapeutically:

  • Context-specific targeting strategies:

    • In ovarian cancer: Inhibitors of TMEM88 could potentially resensitize tumors to platinum-based chemotherapy

    • In thyroid cancer: Activators of TMEM88 expression or function might suppress tumor growth

    • Delivery systems that target specific subcellular compartments based on the cancer type

  • Structural biology approaches:

    • Determination of the three-dimensional and crystal structure of TMEM88 to enable structure-based drug design

    • Focus on the Val-Trp-Val motif that mediates PDZ domain interactions with Dvl

    • Development of peptidomimetics that could modulate TMEM88-Dvl interactions

  • Epigenetic modulation strategies:

    • In cancers where TMEM88 is silenced by hypermethylation (like NSCLC), targeted demethylating approaches

    • Combination of demethylating agents with other therapies for synergistic effects

  • Antibody-based therapeutics:

    • Following the successful model of Claudin18.2-targeting therapies like zolbetuximab

    • Development of antibody-drug conjugates (ADCs) targeting TMEM88 in cancers where it is overexpressed

    • Bispecific antibodies linking TMEM88 recognition with immune cell recruitment

  • Pathway-based combination approaches:

    • Combining TMEM88 modulation with Wnt pathway inhibitors for synergistic effects

    • Rational design of combination therapies based on TMEM88's context-dependent functions

The development of these approaches requires overcoming current limitations, including insufficient structural information and limited understanding of TMEM88's full-length protein polypeptide and recognition epitopes .

How might advanced genetic engineering approaches enhance our understanding of TMEM88 biology?

Advanced genetic engineering techniques offer powerful new approaches to study TMEM88:

  • CRISPR-Cas9 genome editing applications:

    • Generation of endogenously tagged TMEM88 (with fluorescent proteins or affinity tags) to study physiological expression levels

    • Domain-specific mutations to dissect functional regions (e.g., transmembrane domains versus PDZ-binding motif)

    • Tissue-specific conditional knockout models to avoid developmental compensation

  • Single-cell analysis technologies:

    • Single-cell RNA-seq to identify cell populations with differential TMEM88 expression

    • Spatial transcriptomics to map TMEM88 expression patterns within complex tissues

    • Combined with lineage tracing to follow TMEM88 expression through developmental processes

  • Optogenetic and chemogenetic control:

    • Development of light-activated TMEM88 variants to achieve temporal control over its function

    • Chemically inducible dimerization systems to control TMEM88 interactions with binding partners

    • These approaches allow precise manipulation of TMEM88 function in specific subcellular compartments

  • Protein engineering approaches:

    • Creation of biosensors that report on TMEM88 conformational changes or binding events

    • Split-protein complementation assays to visualize TMEM88 interactions in living cells

    • Engineered TMEM88 variants with altered trafficking patterns to distinguish location-specific functions

  • Systems-level screening:

    • Genome-wide CRISPR screens to identify synthetic lethal interactions with TMEM88 in cancer contexts

    • Proteomic approaches like BioID or APEX proximity labeling to map the complete TMEM88 interactome

    • Metabolomic analysis following TMEM88 manipulation to understand broader metabolic consequences

These advanced approaches can provide unprecedented insights into TMEM88 biology that go beyond traditional methods, potentially revealing new functions and therapeutic opportunities.

What methodological challenges remain in translating TMEM88 research findings between in vitro systems and clinical applications?

Several methodological challenges must be addressed to effectively translate TMEM88 research findings:

  • Model system limitations:

    • Cell lines may not recapitulate the complex tissue environment and three-dimensional architecture

    • Patient-derived organoids or xenografts may better preserve tissue-specific TMEM88 functions

    • Development of physiologically relevant 3D culture systems incorporating stromal components

  • Technical standardization needs:

    • Standardized immunohistochemical protocols for consistent TMEM88 detection in clinical samples

    • Validated scoring systems for nuclear versus cytoplasmic TMEM88 expression

    • Development of companion diagnostics to identify patients likely to respond to TMEM88-targeted therapies

  • Mechanistic understanding gaps:

    • Deeper characterization of post-translational modifications affecting TMEM88 function

    • Better understanding of isoform-specific functions in different tissues

    • Clarification of conflicting findings regarding TMEM88's role in specific cancer types

  • Translational research priorities:

    • Correlation of TMEM88 expression/localization patterns with clinical outcomes across larger patient cohorts

    • Development of biomarker panels combining TMEM88 with other prognostic markers

    • Identification of pharmacologically targetable nodes in TMEM88-related pathways

  • Drug development challenges:

    • Design of high-throughput screening assays for compounds that modulate TMEM88 function

    • Development of delivery systems that can target specific subcellular pools of TMEM88

    • Creation of therapeutic approaches that can distinguish between beneficial and harmful TMEM88 functions

Addressing these methodological challenges will be essential for effectively translating the growing body of TMEM88 research into clinical applications that improve patient outcomes.

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 2025 TheBiotek. All Rights Reserved.