BTBD10 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

The BTBD10 antibody (Catalog No. 16995-1-AP) is a rabbit polyclonal antibody developed by Proteintech for research applications targeting the BTBD10 protein. BTBD10, a BTB/POZ domain-containing protein, has been implicated in Akt signaling regulation, motor neuron survival, and cancer progression. This antibody is validated for use in ELISA and demonstrates reactivity with human, mouse, and rat samples, making it a versatile tool for cross-species studies.

Applications

The BTBD10 antibody is optimized for ELISA and has potential utility in additional assays such as western blotting, immunoprecipitation, or immunohistochemistry, pending validation. Its specificity for BTBD10 enables researchers to study:

  • Akt signaling pathways in motor neuron survival (e.g., ALS models) .

  • Tumor biology, including glioma and hepatocellular carcinoma (HCC) , where BTBD10 expression correlates with disease progression.

Research Relevance

BTBD10’s role in Akt activation (via inhibition of PP2A-mediated dephosphorylation) positions it as a critical regulator of cell survival and proliferation . This antibody facilitates:

  • Prognostic biomarker studies in HCC, where high BTBD10 expression predicts poor survival .

  • Therapeutic target validation, particularly in immunotherapy contexts where BTBD10 modulates tumor immune evasion .

Validation Data

Proteintech’s antibody has undergone rigorous testing:

  • ELISA: Demonstrated specificity for BTBD10 in human, mouse, and rat lysates.

  • Western blot: Preliminary data suggest compatibility, pending further optimization.

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and location. For specific delivery timeframes, please consult your local distributor.
Synonyms
BTBD10 antibody; GMRP1BTB/POZ domain-containing protein 10 antibody; Glucose metabolism-related protein 1 antibody
Target Names
BTBD10
Uniprot No.

Target Background

Function
BTBD10 plays a crucial role in activating AKT family members by inhibiting PPP2CA-mediated dephosphorylation, thereby maintaining AKTs in an active state. This function contributes to the prevention of motor neuronal death and the promotion of pancreatic beta cell growth.
Gene References Into Functions
  1. Research indicates that reduced BTBD10 expression is strongly associated with the development of sporadic amyotrophic lateral sclerosis. PMID: 23320755
  2. Collectively, these findings suggest that decreased BTBD10 expression leads to motor neuron death both in vitro and in vivo. PMID: 22388351
  3. GMRP1 regulates pancreatic beta cell proliferation and apoptosis through the activation of the Akt signaling pathway. PMID: 21267538
  4. BTBD10 appears to act as a suppressor of cell death, including neuronal cell death associated with amyotrophic lateral sclerosis, and an enhancer of cell growth through its positive regulation of Akt phosphorylation. PMID: 18160256
Database Links

HGNC: 21445

OMIM: 615933

KEGG: hsa:84280

STRING: 9606.ENSP00000278174

UniGene: Hs.332382

Subcellular Location
Nucleus. Cytoplasm.
Tissue Specificity
Ubiquitously expressed. Highly expressed in adult brain, testis, aorta and small intestine and weakly expressed in the heart, lung, liver, kidney, pancreas, spleen, thymus, prostate, ovary and colon. Down-regulated in glioma.

Q&A

What is BTBD10 and what cellular functions does it regulate?

BTBD10 (BTB/POZ domain-containing protein 10) is a novel Akt-interacting protein that functions as an activator of Akt family members. It operates by decreasing protein phosphatase 2A-mediated dephosphorylation of Akt, thereby enhancing Akt signaling pathway activity. BTBD10 appears to function as a suppressor of cell death, including neuronal cell death related to amyotrophic lateral sclerosis (ALS), and as an enhancer of cell growth through its positive regulation of Akt phosphorylation . The protein is expressed ubiquitously in vivo and distributes in cells as a unique cytoplasmic filamentous structure concentrated around the nucleus . At the molecular level, BTBD10 contains a BTB/POZ domain that mediates protein-protein interactions, allowing it to bind equally to any Akt isoform and regulate downstream signaling cascades.

What are the basic characteristics of commercially available BTBD10 antibodies?

The typical BTBD10 antibody used in research applications has the following characteristics:

PropertySpecification
ReactivityHuman, mouse, rat
Host/IsotypeRabbit/IgG
ClassPolyclonal
ApplicationsELISA, Western blot, IHC
ImmunogenBTBD10 fusion protein
Target ProteinBTB (POZ) domain containing 10
Molecular Weight475 aa, 54 kDa
Storage Conditions-20°C in PBS with 0.02% sodium azide and 50% glycerol (pH 7.3)

The antibody is typically purified using antigen affinity methods and demonstrates consistent reactivity across multiple mammalian species, making it versatile for comparative studies across model organisms .

How should researchers optimize Western blot protocols for BTBD10 detection?

For optimal Western blot detection of BTBD10, researchers should consider the following methodological approach:

  • Sample preparation: Extract proteins using RIPA buffer supplemented with protease and phosphatase inhibitors to preserve both total BTBD10 and its phosphorylated forms.

  • Gel selection: Use 10% SDS-PAGE gels for optimal separation around the 54 kDa range where BTBD10 migrates.

  • Transfer conditions: Employ semi-dry transfer at 15V for 60 minutes or wet transfer at 100V for 90 minutes using PVDF membranes (0.45 μm pore size) for best protein retention.

  • Blocking: Block membranes with 5% non-fat milk in TBST for 1 hour at room temperature to minimize background.

  • Primary antibody incubation: Dilute BTBD10 antibody 1:500-1:1000 in blocking buffer and incubate overnight at 4°C.

  • Detection method: Use HRP-conjugated secondary antibodies with enhanced chemiluminescence for standard detection. For quantitative analysis, consider fluorescent secondary antibodies with an infrared imaging system.

  • Controls: Always include positive controls (tissues known to express BTBD10) and negative controls (BTBD10 knockdown samples) to validate antibody specificity .

When troubleshooting, consider that BTBD10 forms cytoplasmic filamentous structures, which may require specialized lysis conditions to fully solubilize the protein for consistent detection.

How can researchers effectively investigate BTBD10's role in regulating Akt signaling pathways?

To investigate BTBD10's regulatory effect on Akt signaling, researchers should implement a multi-faceted experimental approach:

  • Co-immunoprecipitation assays: Use BTBD10 antibodies to pull down protein complexes and probe for Akt family members and protein phosphatase 2A (PP2A) components to confirm physical interactions. Reciprocal co-IPs with Akt antibodies can verify binding specificity.

  • Phosphorylation analysis: Monitor Akt phosphorylation at Ser473 and Thr308 following BTBD10 modulation (overexpression or knockdown) using phospho-specific antibodies. This should be performed under both basal conditions and following stimulation with growth factors.

  • PP2A activity assays: Measure PP2A phosphatase activity in the presence and absence of BTBD10 using commercial phosphatase assay kits to quantify BTBD10's inhibitory effect on PP2A-mediated Akt dephosphorylation.

  • Downstream target validation: Assess phosphorylation status of known Akt substrates (e.g., GSK3β, FOXO, mTOR) to confirm functional consequences of BTBD10-mediated Akt activation.

  • Domain mapping experiments: Generate BTBD10 truncation mutants to identify which domains are essential for Akt binding and activation versus PP2A inhibition .

When interpreting results, researchers should consider that BTBD10's effects may vary across different cell types and physiological contexts, necessitating validation in multiple experimental systems.

What methodologies should be employed to study BTBD10 in the context of neurodegeneration?

To study BTBD10 in neurodegeneration contexts, particularly in ALS, researchers should consider these methodological approaches:

  • Neuronal culture systems:

    • Primary motor neuron cultures from wild-type and ALS model mice (e.g., G93A-SOD1 transgenic)

    • iPSC-derived motor neurons from ALS patients and healthy controls

    • Apply survival assays following BTBD10 modulation via viral vectors or siRNA

  • In vivo models:

    • Generate BTBD10 knockout or conditional knockout mice

    • Cross with established ALS models (SOD1-G93A, TDP-43, FUS)

    • Perform behavioral testing (rotarod, grip strength, gait analysis)

    • Histopathological examination of spinal cord motor neurons

  • C. elegans models:

    • Utilize btbd-10 gene disruption strains to assess neuronal integrity and locomotion

    • Perform rescue experiments with wild-type human BTBD10

    • Combine with ALS-related transgenes to evaluate genetic interactions

  • Molecular analyses:

    • Quantify BTBD10 expression in post-mortem tissue from ALS patients

    • Assess correlation between BTBD10 levels and disease progression

    • Investigate potential post-translational modifications affecting BTBD10 function

When designing these experiments, it's crucial to distinguish between BTBD10's direct neuroprotective effects and indirect effects mediated through Akt pathway modulation by including appropriate Akt inhibitor controls.

How does BTBD10 expression correlate with cancer prognosis and what experimental approaches can verify its clinical significance?

BTBD10's role as a prognostic biomarker in cancer, particularly hepatocellular carcinoma (HCC), can be investigated through these methodological approaches:

  • Clinical correlation studies:

    • Perform immunohistochemistry on tissue microarrays containing tumor and adjacent normal tissues

    • Conduct BTBD10 expression analysis in large patient cohorts with complete clinical follow-up data

    • Apply multivariate Cox regression analysis to determine if BTBD10 is an independent prognostic factor

  • Mechanistic investigations:

    • Assess relationship between BTBD10 expression and tumor-infiltrating lymphocytes (TILs) using multiplexed immunofluorescence

    • Analyze correlation with immune checkpoint molecules (PD-1, PD-L1, CTLA4) using co-expression studies

    • Investigate association with m6A methylation and ferroptosis pathways through gene expression correlation analyses

  • Functional validation:

    • Modulate BTBD10 expression in cancer cell lines and assess:

      • Proliferation and survival

      • Migration and invasion capacity

      • Immune cell co-culture responses

    • Use xenograft models with BTBD10 overexpression or knockdown to evaluate tumor growth and immune infiltration in vivo

  • Therapeutic response prediction:

    • Correlate BTBD10 expression levels with IC50 values for various targeted therapies

    • Perform combination studies with immune checkpoint inhibitors and BTBD10 modulation

What are the optimal immunohistochemistry protocols for BTBD10 detection in different tissue types?

For effective immunohistochemical detection of BTBD10 across different tissue types, researchers should follow these optimized protocols:

  • Tissue preparation:

    • Fix tissues in 10% neutral-buffered formalin for 24-48 hours

    • Process and embed in paraffin using standard protocols

    • Section at 4-5 μm thickness onto adhesive slides

  • Antigen retrieval methods (tissue-specific optimization):

    • For liver tissues: Heat-induced epitope retrieval using citrate buffer (pH 6.0) for 20 minutes

    • For nervous system tissues: EDTA buffer (pH 9.0) often yields better results

    • For muscle samples: A combination of enzymatic (proteinase K) and heat retrieval may be necessary

  • Blocking and antibody incubation:

    • Block endogenous peroxidase with 3% hydrogen peroxide

    • Apply protein block (5% normal goat serum) for 30 minutes

    • Incubate with primary BTBD10 antibody (1:100-1:200 dilution) overnight at 4°C

    • For detection, use polymer-based detection systems rather than avidin-biotin to reduce background

  • Counterstaining and evaluation:

    • Counterstain with hematoxylin for 1-2 minutes

    • Assess cytoplasmic staining pattern, noting the characteristic filamentous structures around the nucleus

    • Score based on both intensity (0-3) and percentage of positive cells

  • Controls and validation:

    • Include known positive tissues (liver, spinal cord)

    • Use BTBD10 knockdown tissues as negative controls

    • Perform parallel staining with a second BTBD10 antibody targeting a different epitope for validation

When interpreting results, pay particular attention to the subcellular localization of BTBD10, as its distribution in cytoplasmic filamentous structures around the nucleus is characteristic and important for functional analysis.

How can researchers effectively design BTBD10 knockdown or overexpression experiments?

For rigorous BTBD10 gain- and loss-of-function studies, researchers should implement the following methodological approaches:

  • siRNA/shRNA-mediated knockdown:

    • Design multiple siRNA sequences targeting different regions of BTBD10 mRNA

    • Recommended target regions: nucleotides 250-270, 780-800, and 1200-1220 of the coding sequence

    • Validate knockdown efficiency via qRT-PCR and Western blot

    • Use scrambled siRNA sequences as controls

    • For stable knockdown, utilize lentiviral shRNA constructs with selection markers

  • CRISPR/Cas9-mediated knockout:

    • Design 2-3 guide RNAs targeting early exons of BTBD10

    • Screen clones by genomic PCR, sequencing, and Western blot

    • Generate heterozygous and homozygous knockout lines for dose-dependency studies

    • Create conditional knockout systems (e.g., Cre-loxP) for tissue-specific deletion

  • Overexpression strategies:

    • Clone full-length human BTBD10 cDNA into appropriate vectors

    • For transient expression: use CMV promoter-driven constructs

    • For stable expression: use lentiviral or retroviral systems with selection markers

    • Include epitope tags (FLAG, HA, V5) at C-terminus to avoid interference with N-terminal functional domains

    • Generate domain mutants lacking BTB/POZ domain to serve as functional controls

  • Rescue experiments:

    • In knockdown/knockout systems, reintroduce wild-type or mutant BTBD10 constructs

    • Use constructs resistant to siRNA (by introducing silent mutations) for specific rescue

    • Quantify restoration of phenotypes (cell survival, Akt phosphorylation)

When conducting these experiments, it's critical to validate findings across multiple cell types and to examine time-dependent effects, as BTBD10's impact on signaling pathways may vary temporally following modulation.

What analytical approaches should be used to investigate BTBD10's relationship with tumor-infiltrating lymphocytes (TILs)?

To comprehensively analyze BTBD10's relationship with tumor-infiltrating lymphocytes, researchers should employ these analytical approaches:

  • Bioinformatic analyses:

    • Utilize tools like TIMER (Tumor Immune Estimation Resource) to correlate BTBD10 expression with immune cell infiltration

    • Perform GSEA (Gene Set Enrichment Analysis) to identify immune-related pathways associated with BTBD10 expression

    • Apply deconvolution algorithms (e.g., CIBERSORT, xCell) to estimate immune cell type proportions from bulk RNA-seq data

  • Experimental validation techniques:

    • Multiplexed immunofluorescence to simultaneously detect BTBD10 and immune cell markers

    • Flow cytometry on dissociated tumors to quantify immune populations based on BTBD10 expression levels

    • Single-cell RNA sequencing to define cell-specific expression patterns and intercellular communication

  • Functional studies:

    • Co-culture experiments with BTBD10-modulated tumor cells and immune cells

    • Measure cytokine production, proliferation, and cytotoxic activity

    • Assess immune checkpoint molecule expression (PD-1, PD-L1, CTLA4) following BTBD10 modulation

  • In vivo immune monitoring:

    • Implant BTBD10-overexpressing or knockdown tumor cells in immunocompetent mice

    • Analyze tumor growth, immune infiltration, and response to immunotherapies

    • Perform adoptive transfer experiments to assess specific immune cell contributions

Research has demonstrated that BTBD10 expression correlates positively with the presence of various immune cell populations including B cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells in HCC. Additionally, BTBD10 expression correlates with immune checkpoint molecules, suggesting it may influence tumor immune evasion mechanisms .

How does BTBD10 expression change in neurodegenerative disease models and what are the functional consequences?

BTBD10 expression undergoes significant changes in neurodegenerative disease contexts, particularly in ALS models, with important functional implications:

  • Expression patterns in ALS:

    • Decreased BTBD10 expression has been observed in motor neurons from:

      • Sporadic ALS patients' post-mortem tissues

      • G93A-SOD1 transgenic mice (ALS model)

    • This reduction appears to precede motor neuron degeneration, suggesting it may be an early pathogenic event rather than a consequence of neuronal damage

  • Functional consequences of reduced expression:

    • siRNA-mediated inhibition of BTBD10 in cultured motor neurons leads to neuronal death

    • In C. elegans models, disruption of the btbd-10 gene causes:

      • Loss of both motor and touch-receptor neurons

      • Significant locomotion defects

      • These phenotypes occur independent of other ALS-associated mutations

  • Neuroprotective effects of BTBD10 restoration:

    • Overexpression of BTBD10 suppresses motor neuron death induced by the G93A-SOD1 mutant in vitro

    • This protection is mediated through increased Akt phosphorylation and activation

    • The effect is diminished when Akt signaling is pharmacologically blocked

  • Molecular mechanisms:

    • Reduced BTBD10 leads to increased PP2A-mediated dephosphorylation of Akt

    • Decreased Akt activity results in reduced phosphorylation of pro-apoptotic factors

    • This ultimately triggers apoptotic cascades in motor neurons

These findings collectively suggest that BTBD10 reduction represents a common pathway in motor neuron degeneration across different forms of ALS, positioning it as both a potential biomarker and therapeutic target.

What methodological approaches should be used to investigate BTBD10's role in hepatocellular carcinoma prognosis?

To rigorously investigate BTBD10's role in hepatocellular carcinoma (HCC) prognosis, researchers should implement these methodological approaches:

  • Clinical sample analysis:

    • Collect matched tumor and adjacent normal tissues from HCC patients with complete clinical data

    • Perform qRT-PCR and immunohistochemistry to quantify BTBD10 expression

    • Calculate optimal cutoff values for BTBD10 expression using ROC curve analysis

    • Conduct Kaplan-Meier survival analyses and multivariate Cox regression to establish prognostic significance

  • Multi-omics integration:

    • Correlate BTBD10 expression with genomic alterations (mutations, CNVs)

    • Perform proteomics to identify BTBD10 interaction partners in HCC

    • Conduct metabolomic profiling to link BTBD10 expression with metabolic reprogramming

  • Pathway analysis:

    • Investigate BTBD10's relationship with:

      • m6A methylation-related genes (METTL3, METTL14, WTAP, FTO, ALKBH5)

      • Ferroptosis-related genes (NFE2L2, ACSL4, SLC7A11)

      • Immune checkpoint molecules (PD-1, PD-L1, CTLA4)

    • Use GSEA to identify enriched signaling pathways in BTBD10-high vs. BTBD10-low tumors

  • Therapeutic response prediction:

    • Analyze IC50 scores for various targeted therapies based on BTBD10 expression

    • Perform drug sensitivity testing on patient-derived organoids with varying BTBD10 levels

    • Correlate BTBD10 expression with clinical response to sorafenib and immune checkpoint inhibitors

How can researchers design experiments to resolve contradictory findings about BTBD10's role in different cancer types?

To address contradictory findings regarding BTBD10's function across different cancer types, researchers should employ these experimental design strategies:

  • Comprehensive cancer type comparison:

    • Perform parallel analyses of BTBD10 expression and function across multiple cancer types:

      • Use standardized techniques (qRT-PCR, Western blot, IHC) with identical protocols

      • Include paired normal tissues as controls for each cancer type

      • Analyze at least 3-5 cell lines per cancer type to account for heterogeneity

  • Context-dependent signaling analysis:

    • Investigate BTBD10's impact on Akt signaling in different cellular contexts:

      • Compare basal and stimulated Akt phosphorylation levels

      • Assess expression levels of PP2A regulatory subunits that may influence BTBD10-PP2A interaction

      • Examine feedback mechanisms that might counteract BTBD10's effect on Akt in specific tissues

  • Genetic background characterization:

    • Profile key genetic alterations that might interact with BTBD10's function:

      • PTEN status (common Akt pathway regulator)

      • PI3K mutations (upstream of Akt)

      • mTOR pathway alterations (downstream of Akt)

      • Determine how these genetic backgrounds modify BTBD10's effects

  • Tissue-specific interactome mapping:

    • Perform immunoprecipitation followed by mass spectrometry in different cancer types

    • Identify tissue-specific interaction partners that might redirect BTBD10's function

    • Validate key interactions using proximity ligation assays in tissue sections

  • Conditional knockout models:

    • Generate tissue-specific BTBD10 knockout mice (using Cre-loxP system)

    • Induce different cancer types in these models

    • Compare phenotypic outcomes to resolve tissue-specific functions

When interpreting contradictory results, researchers should consider that BTBD10 may play dual roles depending on the cellular context: in neuronal cells, it appears protective against cell death, while in certain cancers like HCC, it may promote tumor progression through immune modulation. These apparently opposing functions may reflect tissue-specific roles of the Akt signaling pathway itself, which can promote either survival or proliferation depending on the cellular context.

What are the optimal approaches for studying BTBD10's interaction with the Akt signaling pathway?

To comprehensively characterize BTBD10's interaction with the Akt signaling pathway, researchers should implement these advanced technical approaches:

  • Protein-protein interaction studies:

    • Bimolecular Fluorescence Complementation (BiFC) to visualize BTBD10-Akt interactions in living cells

    • Proximity Ligation Assay (PLA) to detect endogenous interactions in fixed cells/tissues

    • FRET/FLIM analysis to measure interaction dynamics and conformational changes

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction interfaces

  • Live-cell imaging techniques:

    • Generate fluorescent protein-tagged constructs (BTBD10-GFP, Akt-RFP)

    • Perform time-lapse microscopy following stimulation with growth factors

    • Use FRAP (Fluorescence Recovery After Photobleaching) to assess dynamics of BTBD10-Akt complexes

    • Implement optogenetic approaches to temporally control BTBD10 activity

  • Biochemical pathway dissection:

    • In vitro reconstitution assays with purified components (BTBD10, Akt, PP2A)

    • Measure PP2A activity using phosphatase assays with synthetic peptide substrates

    • Perform in vitro kinase assays to assess Akt activity following BTBD10 addition

    • Use phospho-specific antibodies to monitor multiple Akt substrates simultaneously

  • Structural biology approaches:

    • Determine crystal structure of BTBD10-Akt complex

    • Use cryo-EM for larger complexes including PP2A components

    • Perform molecular dynamics simulations to predict conformational changes

    • Design rational mutations based on structural data to disrupt specific interactions

When implementing these approaches, researchers should consider the temporal dynamics of BTBD10-mediated Akt regulation, as the interaction may be transient or stimulus-dependent. Additionally, the subcellular localization of these interactions should be carefully mapped, as BTBD10's unique filamentous distribution around the nucleus may create spatially restricted signaling microdomains.

How can researchers integrate multi-omics data to understand BTBD10's role in disease pathology?

For comprehensive multi-omics integration to elucidate BTBD10's role in disease pathology, researchers should implement these methodological strategies:

  • Multi-level data collection:

    • Transcriptomics: RNA-seq to identify BTBD10-associated gene expression signatures

    • Proteomics: Mass spectrometry to map the BTBD10 interactome and phosphoproteome

    • Epigenomics: ChIP-seq to identify transcription factors regulating BTBD10 expression

    • Metabolomics: LC-MS to detect metabolic alterations associated with BTBD10 modulation

  • Integrative computational approaches:

    • Apply supervised machine learning to identify BTBD10-associated multi-omics signatures

    • Use network-based methods to construct BTBD10-centered molecular interaction networks

    • Implement Bayesian approaches to infer causal relationships between BTBD10 and disease phenotypes

    • Perform joint pathway enrichment across multiple data types

  • Single-cell multi-omics:

    • Conduct single-cell RNA-seq with CITE-seq for simultaneous protein detection

    • Apply spatial transcriptomics to map BTBD10 expression in tissue context

    • Use scATAC-seq to correlate chromatin accessibility with BTBD10 expression

    • Implement trajectory analysis to determine temporal dynamics of BTBD10 function

  • Validation in human biospecimens:

    • Create multi-omics datasets from patient cohorts with varying disease stages

    • Correlate BTBD10 levels with clinical parameters and multi-omics signatures

    • Implement tissue microarrays for high-throughput validation

    • Develop predictive models incorporating BTBD10-related features

This integrative approach revealed that in HCC, BTBD10 expression positively correlates with several m6A methylation-related genes (METTL3, METTL14, WTAP, FTO) and ferroptosis-related genes (NFE2L2, ACSL4, SLC7A11). These correlations provide new perspectives for understanding BTBD10's broader impact on cellular processes beyond Akt signaling and may explain its diverse roles in different pathological contexts .

What are the most sensitive methods for detecting subtle changes in BTBD10 expression and their functional implications?

To detect subtle changes in BTBD10 expression and understand their functional consequences, researchers should employ these highly sensitive methodological approaches:

  • Advanced quantitative expression analysis:

    • Droplet Digital PCR (ddPCR) for absolute quantification of BTBD10 transcript levels

    • NanoString nCounter technology for direct digital counting without amplification bias

    • Single-molecule RNA FISH to visualize individual BTBD10 mRNA molecules in situ

    • Targeted proteomics using Selected Reaction Monitoring (SRM) or Parallel Reaction Monitoring (PRM)

  • High-sensitivity protein detection:

    • Ultrasensitive ELISA with amplification steps (e.g., DELFIA, Simoa)

    • Single-molecule imaging techniques to quantify individual BTBD10 protein molecules

    • Proximity Extension Assay (PEA) for highly specific protein quantification in small samples

    • Capillary Western immunoassay (Wes) for automated, reproducible quantification with minimal sample input

  • Functional readout systems:

    • FRET-based biosensors for Akt activity to monitor immediate consequences of BTBD10 modulation

    • Live-cell reporters for PP2A activity using fluorescent substrates

    • High-content imaging to quantify multiple downstream effects simultaneously

    • Seahorse metabolic flux analysis to detect subtle bioenergetic changes

  • Cell-type specific analysis in heterogeneous samples:

    • Laser Capture Microdissection to isolate specific cell populations

    • Cell sorting based on surface markers followed by BTBD10 analysis

    • Single-cell Western blotting to measure BTBD10 at individual cell level

    • Mass cytometry (CyTOF) with intracellular BTBD10 staining

Research has shown that even subtle reductions in BTBD10 expression can have significant functional impacts. In neurons, partial knockdown of BTBD10 is sufficient to increase vulnerability to stress, while in cancer cells, modest increases in expression can alter immune cell infiltration patterns. These observations highlight the importance of using highly sensitive detection methods that can reliably quantify small but functionally relevant changes in BTBD10 levels.

How can BTBD10 be therapeutically targeted in neurodegenerative diseases and what preclinical models would be most appropriate?

For developing therapeutic strategies targeting BTBD10 in neurodegenerative diseases, researchers should consider these methodological approaches and preclinical models:

  • Therapeutic approaches for BTBD10 modulation:

    • Gene therapy strategies:

      • AAV9-mediated BTBD10 overexpression (crosses blood-brain barrier)

      • Promoter-specific expression in motor neurons using HB9 or ChAT promoters

      • Regulatable expression systems (e.g., Tet-On) for controlled dosing

    • Small molecule development:

      • High-throughput screening for compounds that stabilize BTBD10-Akt interaction

      • Inhibitors of PP2A-BTBD10 interaction to enhance Akt activation

      • Compounds that increase endogenous BTBD10 expression

    • RNA-based therapies:

      • Antisense oligonucleotides targeting BTBD10 negative regulators

      • mRNA delivery for transient BTBD10 expression

  • Optimal preclinical models:

    • In vitro models:

      • iPSC-derived motor neurons from ALS patients (familial and sporadic)

      • Primary motor neuron cultures from rodents

      • Organoid models incorporating multiple cell types (neurons, glia, vascular)

    • Invertebrate models:

      • C. elegans with btbd-10 mutations combined with ALS-related transgenes

      • Drosophila models with tissue-specific BTBD10 modulation

    • Rodent models:

      • G93A-SOD1 transgenic mice (rapid disease progression)

      • TDP-43 models (slower progression, more relevant to sporadic ALS)

      • BTBD10 knockout mice crossed with ALS models

    • Large animal models:

      • Naturally occurring canine degenerative myelopathy (DM)

      • Minipig models for scaling up therapeutic delivery approaches

When designing therapeutic interventions, timing is critical - studies in ALS models have shown that BTBD10 expression decreases before symptom onset, suggesting that early intervention may be necessary for maximum efficacy. Additionally, combination approaches targeting both BTBD10 and other neuroprotective pathways may provide synergistic benefits in complex neurodegenerative conditions.

What experimental design would best evaluate BTBD10 as an immunotherapy target in hepatocellular carcinoma?

To evaluate BTBD10 as an immunotherapy target in hepatocellular carcinoma (HCC), researchers should implement this comprehensive experimental design:

  • Target validation studies:

    • siRNA/CRISPR-mediated BTBD10 knockdown/knockout in HCC cell lines

    • Assessment of changes in:

      • Immune checkpoint molecule expression (PD-L1, PD-L2, CTLA4)

      • Cytokine/chemokine production profile

      • Antigen presentation machinery

    • Parallel overexpression studies to confirm bidirectional effects

  • In vitro immune interaction models:

    • Co-culture systems with BTBD10-modulated HCC cells and:

      • Tumor-infiltrating lymphocytes isolated from HCC patients

      • PBMCs from healthy donors

      • Individual immune cell populations (T cells, NK cells, macrophages)

    • Measure immune cell activation, proliferation, cytotoxicity, and exhaustion markers

    • 3D spheroid co-cultures to better recapitulate tumor microenvironment

  • In vivo preclinical models:

    • Orthotopic implantation of BTBD10-modulated HCC cells in immunocompetent mice

    • Humanized mouse models with human immune system components

    • Treatment arms:

      • Anti-PD-1/PD-L1 monotherapy

      • BTBD10 targeting (siRNA, small molecules)

      • Combination therapy

      • Control groups

    • Endpoints: tumor growth, survival, immune infiltration profile, metastasis

  • Biomarker development:

    • Identify BTBD10-associated immune signatures predictive of immunotherapy response

    • Develop companion diagnostics for patient stratification

    • Explore liquid biopsy approaches to monitor BTBD10 status during treatment

Research has already established that BTBD10 expression positively correlates with tumor-infiltrating B cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells in HCC. Additionally, BTBD10 expression is synchronized with several immune checkpoints. These findings suggest that modulating BTBD10 could potentially enhance the efficacy of existing immunotherapies by altering the tumor immune microenvironment .

How can researchers design experiments to determine if the IC50 differences in drug response based on BTBD10 expression have clinical relevance?

To establish the clinical relevance of IC50 differences in drug response based on BTBD10 expression, researchers should implement this systematic experimental design:

Research has already demonstrated that the IC50 scores of several drugs, including Sorafenib (the standard first-line treatment for advanced HCC), Navitoclax, Veliparib, Luminespib, and Imatinib, are lower in BTBD10 high-expressing HCC. This suggests that BTBD10 expression may serve as a predictive biomarker for drug sensitivity, potentially allowing for more personalized treatment approaches in HCC management .

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.