MAP4K4 Antibody, Biotin conjugated would consist of:
Primary antibody: Specific to MAP4K4 (mitogen-activated protein kinase kinase kinase kinase 4), a serine/threonine kinase involved in stress response, cell motility, and Hippo/YAP signaling pathways.
Conjugate: Biotin (vitamin B7) covalently linked via lysine or cysteine residues for high-affinity streptavidin binding in detection assays.
| Application | Purpose |
|---|---|
| Western Blot (WB) | Amplified signal detection using streptavidin-HRP or fluorescent streptavidin derivatives. |
| Immunohistochemistry (IHC) | Enhanced sensitivity for low-abundance MAP4K4 in tissue sections. |
| ELISA | Quantification of MAP4K4 in biological samples. |
| Flow Cytometry | Cell-surface or intracellular MAP4K4 detection. |
While no direct studies on biotin-conjugated MAP4K4 antibodies are cited in the provided sources, MAP4K4 itself has been characterized as:
A regulator of tumor invasiveness via ERM protein phosphorylation and Arp2/3-mediated actin remodeling .
A suppressor of YAP/TAZ signaling in medulloblastoma cells under basal conditions .
A stress-responsive kinase interacting with STRIPAK complex components (e.g., STRN3) to modulate growth pathways .
Based on analogous antibody data ( ):
Recommended dilutions:
WB: 1:200–1:2000
IHC: 1:20–1:200
ELISA: Up to 1:10,000
Cross-reactivity: Likely limited to human and mouse based on MAP4K4 homology .
Storage: Stable at -20°C with 50% glycerol; avoid freeze-thaw cycles .
No peer-reviewed studies in the provided sources specifically address biotinylated MAP4K4 antibodies. Current literature focuses on:
MAP4K4 (Mitogen-activated protein kinase kinase kinase kinase 4) is a serine/threonine kinase that functions as a critical regulator of multiple cellular processes. It plays essential roles in responding to environmental stress and cytokines such as TNF-alpha by acting upstream of the JUN N-terminal pathway . MAP4K4 serves as an activator of the Hippo signaling pathway, which controls organ size and tumor suppression by restricting proliferation and promoting apoptosis . It operates in parallel with and partially redundantly to STK3/MST2 and STK4/MST2 in phosphorylating and activating LATS1/2 . Additionally, MAP4K4 phosphorylates SMAD1 on Thr-322, regulating TGF-β family signaling pathways . In cancer progression, MAP4K4 functions as a central regulator of collective cell migration, controlling protrusion dynamics, cytoskeletal remodeling, and adhesion stability . Recent research has also identified MAP4K4 as a negative regulator of CD8 T cell activation through its effects on LFA-1 activation and T cell-APC conjugation .
Biotin conjugation enhances MAP4K4 antibody utility through several methodological advantages. The biotin-streptavidin system provides exceptional sensitivity due to the high-affinity interaction (Kd ≈ 10^-15 M) between biotin and streptavidin/avidin, enabling detection of low-abundance MAP4K4 protein in complex samples . The conjugation allows for signal amplification through secondary detection systems that leverage multiple biotin-binding sites on streptavidin molecules . In multiplex experimental designs, biotin-conjugated MAP4K4 antibodies can be combined with differently labeled primary antibodies to simultaneously detect multiple targets without cross-reactivity issues . The biotin-streptavidin system is also compatible with various detection methods including ELISA, immunohistochemistry, and flow cytometry, providing experimental flexibility . For researchers investigating MAP4K4's role in cell migration or stress response, biotin-conjugated antibodies facilitate precise subcellular localization studies through streptavidin-conjugated quantum dots or fluorophores with minimal steric hindrance .
To maintain optimal activity of biotin-conjugated MAP4K4 antibodies, precise storage and handling protocols must be followed based on manufacturer specifications and research best practices. Upon receipt, store the antibody at -20°C or -80°C to prevent degradation, avoiding repeated freeze-thaw cycles that can diminish binding efficiency and increase background signal . The antibody is typically provided in a stabilizing buffer containing 50% glycerol and 0.01M PBS (pH 7.4) with 0.03% Proclin 300 as a preservative, which helps maintain protein structure and prevents microbial growth . When working with the antibody, maintain cold chain practices by keeping it on ice or at 4°C during experimental procedures . For dilution, use fresh, sterile buffers appropriate for your application (typically PBS with 0.1-0.5% BSA or similar carrier protein) to prevent non-specific binding . Always use DNase/RNase-free laboratory consumables and reagents when handling the antibody to prevent contamination that might interfere with downstream applications . Monitor expiration dates and batch-to-batch variation by maintaining detailed records of antibody performance, particularly in sensitive applications like ELISA .
When optimizing ELISA protocols with biotin-conjugated MAP4K4 antibodies, researchers should implement a systematic approach to maximize sensitivity and specificity. Begin with a titration experiment using 2-fold serial dilutions (typically ranging from 1:100 to 1:12,800) of the biotin-conjugated MAP4K4 antibody against a constant amount of antigen to determine the optimal working concentration that provides maximum signal with minimal background . The optimal coating buffer for the capture antibody is typically 0.05M carbonate-bicarbonate buffer (pH 9.6), while PBS (pH 7.4) with 0.05% Tween-20 and 1-3% BSA serves as an effective blocking buffer to minimize non-specific binding . When developing the detection system, streptavidin-HRP conjugates are recommended at dilutions between 1:1000 and 1:5000, with incubation times of 30-60 minutes at room temperature in a humidified chamber . For enhanced sensitivity in detecting low-abundance MAP4K4 in complex samples such as tumor lysates, implement a sandwich ELISA format with a MAP4K4-specific capture antibody targeting a different epitope than the biotin-conjugated detection antibody . Include multiple technical replicates (at least triplicates) and appropriate controls, including a standard curve using recombinant MAP4K4 protein (866-1104AA region), blank wells (no antigen), and isotype controls to account for non-specific binding . The wash steps are critical - perform at least 3-5 washes between each step using PBS with 0.05% Tween-20, ensuring complete removal of wash buffer between steps to minimize background signal .
When investigating MAP4K4's role in cell migration with biotin-conjugated antibodies, a comprehensive control strategy is essential to ensure experimental validity. Primary controls should include isotype-matched antibodies conjugated to biotin to distinguish between specific binding and Fc receptor-mediated non-specific interactions, particularly in immunofluorescence or flow cytometry applications . For functional studies, include both MAP4K4 knockdown cells (using validated siRNA or CRISPR-Cas9) and MAP4K4 overexpression systems as positive and negative controls to establish the specificity of migration phenotypes . When analyzing collective cell migration in models like A431 carcinoma cells, implement both 2D wound healing assays and 3D invasion assays to comprehensively assess MAP4K4's role in different migration contexts . Time-course experiments are crucial to distinguish immediate versus delayed effects of MAP4K4 inhibition on migration, with recommended timepoints at 0, 6, 12, 24, and 48 hours post-treatment . For mechanistic studies examining MAP4K4's effects on adhesion dynamics, include controls focusing on specific downstream targets such as focal adhesion proteins and adherens junction components, comparing phosphorylation states in the presence and absence of MAP4K4 activity . When analyzing biomechanical forces using traction force microscopy, implement both substrate stiffness controls (varying from 2-50 kPa) and cell density controls to account for these variables' effects on force generation and transmission .
Validating MAP4K4 antibody specificity requires a multi-technique approach to ensure reliable experimental outcomes. Western blot analysis should be performed using both recombinant MAP4K4 protein and endogenous MAP4K4 from cellular lysates, confirming a single band at the expected molecular weight of approximately 142-171 kDa depending on post-translational modifications . Genetic validation through MAP4K4 knockdown/knockout systems is essential, with siRNA treatment showing corresponding reduction in antibody signal intensity; this approach has been successfully demonstrated in multiple cell types including HeLa cells and endothelial cells . Immunoprecipitation followed by mass spectrometry can provide definitive confirmation that the antibody captures the intended target—this technique has shown that anti-MAP4K4 antibodies can successfully pull down MAP4K4 from complex cell lysates with high specificity . Cross-reactivity testing should be performed against closely related kinases in the MAP4K family (MAP4K1-3, MAP4K5) to ensure signal specificity, as these proteins share structural similarities that could lead to misinterpretation of results . For immunofluorescence applications, co-staining with alternative MAP4K4 antibodies targeting different epitopes should show co-localization patterns; additionally, fluorescence signal should correlate with known expression patterns of MAP4K4 in tissues where it has been well-characterized, such as in immune cells or tumor tissues . Lot-to-lot consistency should be evaluated through comparative testing of multiple antibody batches using standardized protein samples to ensure reproducibility across experiments .
Biotin-conjugated MAP4K4 antibodies serve as powerful tools for investigating MAP4K4's regulatory role in CD8 T cell antitumor immunity through several advanced methodological approaches. Flow cytometry-based phospho-protein analysis allows researchers to quantitatively assess MAP4K4-dependent phosphorylation of ezrin, radixin, and moesin (ERM) proteins in CD8 T cells following activation stimuli, revealing how MAP4K4 dynamically regulates these cytoskeletal linkers during immune synapse formation . Proximity ligation assays utilizing biotin-conjugated MAP4K4 antibodies in conjunction with LFA-1 antibodies can directly visualize molecular associations between MAP4K4 and the integrin machinery at immunological synapses, providing spatial resolution below 40nm of these crucial regulatory interactions . For mechanistic studies of T cell-APC interactions, researchers can employ microfluidic-based single-cell force spectroscopy with MAP4K4 antibody-coated surfaces to measure binding strength differences between wild-type and MAP4K4-deficient CD8 T cells, quantifying how MAP4K4 modulates adhesion forces during T cell priming . In adoptive T cell therapy models, biotin-conjugated MAP4K4 antibodies facilitate tracking of MAP4K4 protein levels in engineered T cells via flow cytometry before and after tumor infiltration, correlating MAP4K4 expression with antitumor efficacy . Multi-parameter confocal imaging of tumor-infiltrating lymphocytes using biotin-conjugated MAP4K4 antibodies with streptavidin-fluorophore detection systems can reveal spatiotemporal dynamics of MAP4K4 activation in response to the tumor microenvironment, particularly in relationship to exhaustion markers and functional cytokine production . This approach has demonstrated that genetic deletion of Map4k4 increases CD8 T cell priming, which culminates in enhanced antigen-dependent activation, proliferation, cytokine production, and cytotoxic activity, resulting in impaired tumor growth .
To study MAP4K4's role in regulating biomechanical forces during collective cell migration, researchers should implement a multi-modal methodological framework that integrates advanced biophysical techniques with molecular biology approaches. Traction force microscopy with fluorescent microbeads embedded in polyacrylamide gels of defined stiffness (typically 2-12 kPa) allows quantification of cell-substrate forces in both MAP4K4-expressing and MAP4K4-depleted cell clusters, revealing how MAP4K4 modulates force generation and distribution across migrating collectives . These measurements have demonstrated that MAP4K4 loss of function leads to tensional disequilibrium throughout cell clusters, increasing traction forces at cell-cell adhesions . Monolayer stress microscopy, which computationally infers intercellular stresses from traction force measurements, provides insights into how MAP4K4 regulates force transmission between neighboring cells in epithelial sheets . FRET-based tension sensors incorporated into key adhesion molecules (E-cadherin, vinculin) enable real-time visualization of molecular tension within individual adhesion complexes, revealing how MAP4K4 dynamically regulates these mechanical linkages during collective migration . Time-lapse phase contrast microscopy coupled with particle image velocimetry analysis quantifies velocity fields and deformation patterns in migrating cell sheets, demonstrating how MAP4K4 coordinates protrusion and retraction dynamics across multiple cells . Atomic force microscopy in force spectroscopy mode provides direct measurements of cell-cell adhesion strength in control versus MAP4K4-depleted cells, quantifying how MAP4K4 activity modulates intercellular cohesion . To connect mechanical measurements with molecular mechanisms, researchers should perform phosphoproteomics following MAP4K4 perturbation to identify mechanosensitive targets, particularly focusing on adhesion components and cytoskeletal regulators . This comprehensive approach has revealed that MAP4K4 promotes focal adhesion disassembly through phosphorylation of the actin and plasma membrane crosslinker moesin while disassembling adherens junctions through a moesin-independent mechanism .
Investigating MAP4K4's role in KSHV reactivation requires integration of virological, molecular, and cellular approaches to delineate mechanistic pathways. Researchers should establish inducible MAP4K4 knockdown systems in KSHV-infected endothelial cells using doxycycline-regulated shRNA or CRISPR-Cas9 to precisely control MAP4K4 expression during different phases of viral reactivation . Real-time qPCR analysis of viral lytic gene expression (particularly focusing on immediate-early genes like RTA, early genes like KbZIP, and late genes like K8.1) following chemical induction with sodium butyrate in the presence or absence of MAP4K4 reveals the specific stage at which MAP4K4 influences the lytic cascade . This approach has demonstrated that MAP4K4 depletion significantly reduces viral titre, lytic protein expression, and replication, suggesting this kinase contributes to successful completion of the KSHV lytic programme . Viral replication assays using fluorescently labeled KSHV (such as rKSHV.219) combined with flow cytometry and fluorescence microscopy provide quantitative assessment of how MAP4K4 manipulation affects viral production and spread . Transcriptome profiling (RNA-Seq) comparing MAP4K4-silenced versus control cells during KSHV reactivation has identified 54 cellular genes whose expression decreases by at least 1.5-fold after MAP4K4 knockdown, including COX-2, MMP-7, and MMP-13, which are involved in inflammation and invasiveness . Chromatin immunoprecipitation assays can determine whether MAP4K4 regulates transcription factors that bind to viral promoters or cellular genes involved in viral reactivation . For translational relevance, immunohistochemistry using anti-MAP4K4 antibodies on Kaposi's sarcoma tissue specimens can correlate MAP4K4 expression with disease progression and viral activity; this approach has revealed strong MAP4K4 expression in KSHV-infected endothelial spindle cells in KS tissue . Functional invasion assays (transwell or 3D matrix invasion) comparing MAP4K4-depleted versus control KSHV-infected cells quantify the impact on invasive potential, with MAP4K4 knockdown significantly reducing invasion, establishing a mechanistic link between viral pathogenesis and metastatic potential .
Researchers frequently encounter several technical challenges when working with biotin-conjugated MAP4K4 antibodies that require specific troubleshooting approaches. High background signal in immunoassays often stems from endogenous biotin in biological samples, particularly when using avidin-based detection systems . This can be mitigated by implementing a biotin blocking step using commercial biotin blocking kits or by pretreating samples with streptavidin followed by free biotin before applying the biotin-conjugated MAP4K4 antibody . Signal variability between experiments may indicate antibody degradation from improper storage; maintain consistent aliquoting practices upon receipt (typically 10-20μL per aliquot) to minimize freeze-thaw cycles, and store at recommended temperatures (-20°C to -80°C) . When multiple bands appear in Western blot applications, this may represent MAP4K4 isoforms, proteolytic fragments, or non-specific binding . Confirm specificity through MAP4K4 knockdown experiments and optimize blocking conditions (5% BSA is often more effective than milk-based blockers for phospho-specific detection) . Cross-reactivity with related kinases (other MAP4K family members) can complicate interpretation; validate specificity through immunoprecipitation followed by mass spectrometry to confirm target identity . For multiplexing applications where biotin-conjugated MAP4K4 antibodies are used alongside other labeled antibodies, spectral overlap can create false positives; perform single-stain controls with each fluorophore and implement appropriate compensation in flow cytometry or multiplex imaging . Low sensitivity in detecting endogenous MAP4K4 may require signal amplification; employ tyramide signal amplification systems compatible with biotin-streptavidin detection to enhance signal while maintaining specificity .
Distinguishing between total and phosphorylated MAP4K4 requires specialized techniques to accurately assess kinase activation states. Phospho-specific antibodies targeting key regulatory phosphorylation sites on MAP4K4 (particularly Ser801 and Thr202) should be used in parallel with total MAP4K4 antibodies in Western blot analysis, with the phospho/total ratio providing a quantitative measure of activation status . These analyses typically require optimization of sample preparation methods, as rapid dephosphorylation can occur during cell lysis; use phosphatase inhibitor cocktails containing sodium orthovanadate (1-5mM), sodium fluoride (10-50mM), and β-glycerophosphate (10mM) in all buffers, and maintain samples at 4°C throughout processing . Phos-tag SDS-PAGE provides superior resolution of phosphorylated MAP4K4 species without requiring phospho-specific antibodies; this approach incorporates Mn2+-Phos-tag molecules into polyacrylamide gels (typically at 10-100μM concentration), causing mobility shifts proportional to phosphorylation levels that can be detected with total MAP4K4 antibodies . Lambda phosphatase treatment of parallel samples serves as a critical control to confirm phosphorylation-dependent mobility shifts or signal detection . For spatial resolution of activated MAP4K4 in cells, proximity ligation assays combining total MAP4K4 antibodies with phospho-specific antibodies generate fluorescent signals only where both epitopes are in close proximity (typically within 40nm), enabling visualization of activated MAP4K4 pools in specific subcellular compartments . Mass spectrometry-based phosphoproteomics following MAP4K4 immunoprecipitation provides comprehensive mapping of phosphorylation sites with site occupancy quantification, revealing activation patterns that might be missed by antibody-based methods . In MAP4K4-dependent signaling studies, researchers should monitor phosphorylation of known downstream substrates (such as LATS1/2 or SMAD1-Thr322) as proxies for MAP4K4 activity, particularly in contexts where direct MAP4K4 phosphorylation detection is challenging .
MAP4K4 antibodies enable multifaceted investigation of this kinase's role in metabolic disorders through several methodological approaches. Immunohistochemical analysis of adipose tissue biopsies from insulin-resistant versus insulin-sensitive subjects using biotin-conjugated MAP4K4 antibodies can reveal expression patterns correlated with metabolic dysfunction, providing insights into tissue-specific roles . Multi-color flow cytometry with MAP4K4 antibodies combined with markers for macrophage polarization (M1/M2) in adipose tissue can elucidate how MAP4K4 regulates inflammatory responses in obesity, as MAP4K4 is known to mediate TNF-alpha signaling which contributes to insulin resistance . Phosphoproteomic analysis following immunoprecipitation with MAP4K4 antibodies in insulin-responsive tissues (skeletal muscle, liver, adipose) after insulin stimulation can identify novel MAP4K4 substrates involved in glucose homeostasis, enabling the construction of comprehensive signaling networks . For therapeutic development, high-content screening platforms utilizing automated microscopy with MAP4K4 antibodies can assess the effects of small molecule inhibitors on MAP4K4 expression, localization, and downstream signaling in metabolically relevant cell types . Co-immunoprecipitation studies using MAP4K4 antibodies can identify interaction partners specific to metabolic tissues, potentially revealing tissue-specific regulatory mechanisms that could be targeted for therapeutic intervention . MAP4K4 antibodies can also be used in combination with metabolic tracers to correlate MAP4K4 activity with cellular energy utilization, glucose uptake, and lipid metabolism through microscopy or flow cytometry-based approaches . In translational applications, systematic analysis of multiple organs following inducible whole-body deletion of Map4k4 in adult animals has revealed that this intervention is well-tolerated under homeostatic conditions, suggesting potential safety for therapeutic targeting .
When investigating MAP4K4's diverse roles across cancer types using antibodies, researchers must implement contextual methodological strategies that address tissue-specific variations and tumor heterogeneity. Multiplex immunohistochemistry using biotin-conjugated MAP4K4 antibodies alongside markers for tumor subtype, proliferation, and invasion provides spatial context for MAP4K4 expression patterns within the tumor microenvironment; this approach has revealed strong MAP4K4 expression in KSHV-infected endothelial spindle cells in Kaposi's sarcoma tissue . Single-cell Western blot or cytometry by time-of-flight (CyTOF) with MAP4K4 antibodies enables quantification of expression heterogeneity within tumors at the single-cell level, revealing potential resistance mechanisms in MAP4K4-targeted therapies . For mechanistic studies, CRISPR-Cas9 screening combined with MAP4K4 antibody-based detection can identify synthetic lethal interactions specific to MAP4K4-high cancer cells, providing insights into potential combination therapy approaches . Live-cell imaging using fluorescently labeled MAP4K4 antibody fragments in patient-derived xenografts or organoids enables real-time tracking of MAP4K4 dynamics during cancer cell invasion and metastasis, particularly relevant given MAP4K4's role in collective cell migration in A431 carcinoma cells . In drug development applications, pharmacodynamic biomarker studies using MAP4K4 antibodies to detect changes in MAP4K4 expression or activity following treatment with targeted therapies can assess efficacy and resistance mechanisms in patient samples . Cancer type-specific validation is critical, as MAP4K4 functions differently across tumor types; for instance, in carcinoma cells it regulates collective migration through focal adhesion dynamics and adherens junction stability, while in virally-induced cancers it contributes to pathogenesis through regulation of matrix metalloproteinases . When developing predictive biomarkers, quantitative immunohistochemistry or ELISA with carefully validated MAP4K4 antibodies should be correlated with patient outcomes through multivariate analysis accounting for cancer stage, grade, and molecular subtype .
Integrating advanced imaging with MAP4K4 antibodies enables visualization of dynamic kinase regulation with unprecedented spatiotemporal resolution. For intravital imaging, minimally invasive antibody formats such as biotin-conjugated Fab fragments or single-domain antibodies against MAP4K4 conjugated to bright, photostable fluorophores (Alexa647, Janelia Fluor dyes) can penetrate tissues more effectively than full IgG molecules while maintaining specificity . When studying rapid signaling dynamics, researchers can employ genetically encoded FRET-based MAP4K4 activity sensors in combination with antibody-based detection of total MAP4K4 to simultaneously visualize kinase localization and activation state in real-time following stimuli like TNF-alpha or mechanical stress . Super-resolution microscopy techniques including Stimulated Emission Depletion (STED) or Stochastic Optical Reconstruction Microscopy (STORM) with biotin-conjugated MAP4K4 antibodies and streptavidin-fluorophore detection achieve nanoscale resolution (~20-50nm) of MAP4K4 distribution within subcellular structures such as focal adhesions or immune synapses . For tracking MAP4K4 mobility and binding kinetics in living cells, fluorescence recovery after photobleaching (FRAP) or fluorescence correlation spectroscopy (FCS) using fluorescently labeled antibody fragments can measure diffusion rates and binding affinities in different cellular compartments . Lattice light-sheet microscopy combined with adaptive optics enables long-term volumetric imaging of MAP4K4 dynamics in thick samples with minimal phototoxicity, revealing three-dimensional organization during processes like collective cell migration or T cell activation . Correlative light and electron microscopy (CLEM) using biotin-conjugated MAP4K4 antibodies with gold-conjugated streptavidin provides molecular-scale context by correlating fluorescence signals with ultrastructural features, particularly valuable for studying MAP4K4's association with membranes and cytoskeletal elements . For in vivo applications, antibody-based positron emission tomography (immuno-PET) using radiolabeled MAP4K4 antibodies can non-invasively monitor MAP4K4 expression in animal models of cancer or inflammation, providing translational insights into its regulation under physiological conditions .