MTOR Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Storage Buffer: PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Product dispatch typically occurs within 1-3 business days of order receipt. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
dJ576K7.1 (FK506 binding protein 12 rapamycin associated protein 1) antibody; FK506 binding protein 12 rapamycin associated protein 1 antibody; FK506 binding protein 12 rapamycin associated protein 2 antibody; FK506 binding protein 12 rapamycin complex associated protein 1 antibody; FK506-binding protein 12-rapamycin complex-associated protein 1 antibody; FKBP rapamycin associated protein antibody; FKBP12 rapamycin complex associated protein antibody; FKBP12-rapamycin complex-associated protein 1 antibody; FKBP12-rapamycin complex-associated protein antibody; FLJ44809 antibody; FRAP antibody; FRAP1 antibody; FRAP2 antibody; Mammalian target of rapamycin antibody; Mechanistic target of rapamycin antibody; mTOR antibody; MTOR_HUMAN antibody; OTTHUMP00000001983 antibody; RAFT1 antibody; Rapamycin and FKBP12 target 1 antibody; Rapamycin associated protein FRAP2 antibody; Rapamycin target protein 1 antibody; Rapamycin target protein antibody; RAPT1 antibody; Serine/threonine-protein kinase mTOR antibody
Target Names
Uniprot No.

Target Background

Function

The MTOR (mechanistic target of rapamycin) antibody targets a serine/threonine protein kinase that plays a pivotal role in regulating cellular metabolism, growth, and survival. Its activity is modulated by various signals, including hormones, growth factors, nutrients, energy levels, and stress. MTOR directly or indirectly regulates the phosphorylation of at least 800 proteins and functions within two distinct complexes: mTORC1 and mTORC2.

Activated mTORC1 stimulates protein synthesis by phosphorylating key regulators of mRNA translation and ribosome biogenesis. This includes the phosphorylation of EIF4EBP1, releasing its inhibition of the eukaryotic translation initiation factor 4E (eIF4E). Furthermore, mTORC1 phosphorylates and activates RPS6KB1 and RPS6KB2, which further promote protein synthesis by modulating downstream targets such as ribosomal protein S6, eukaryotic translation initiation factor 4B, and the translation initiation inhibitor PDCD4. mTORC1 also controls the activity of the MiT/TFE transcription factors TFEB and TFE3. Under nutrient-rich conditions, it phosphorylates TFEB and TFE3, retaining them in the cytosol and inhibiting their activity. Conversely, under starvation or lysosomal stress, mTORC1 inhibition leads to TFEB and TFE3 dephosphorylation, promoting their nuclear translocation and transcriptional activity.

mTORC1 regulates pyrimidine biosynthesis, both acutely through RPS6KB1-mediated phosphorylation of the biosynthetic enzyme CAD, and chronically via transcriptional enhancement of the pentose phosphate pathway. This pathway produces 5-phosphoribosyl-1-pyrophosphate (PRPP), an allosteric activator of CAD. This function is dependent on the mTORC1 complex. mTORC1 also regulates ribosome synthesis by activating RNA polymerase III-dependent transcription through the phosphorylation and inhibition of MAF1, an RNA polymerase III repressor. In addition to protein synthesis, mTORC1 regulates lipid synthesis through SREBF1/SREBP1 and LPIN1 and, to maintain energy homeostasis, may regulate mitochondrial biogenesis through PPARGC1A. mTORC1 negatively regulates autophagy by phosphorylating ULK1 (at Ser-758) under nutrient-sufficient conditions, disrupting its interaction with AMPK and preventing its activation. It also inhibits autophagy through phosphorylation of the autophagy inhibitor DAP and RUBCNL/Pacer. Furthermore, mTORC1 phosphorylates AMBRA1, thereby inhibiting its ability to mediate ULK1 ubiquitination and the interaction between AMBRA1 and PPP2CA.

mTORC1 exerts feedback control on upstream growth factor signaling, including phosphorylation and activation of GRB10, an insulin receptor (INSR)-dependent signaling suppressor. Other potential mTORC1 targets include CLIP1, which may regulate microtubules.

As part of the mTORC2 complex, MTOR regulates various cellular processes, including cell survival and cytoskeletal organization. A critical role of mTORC2 is the phosphorylation of AKT1 (at Ser-473), a pro-survival effector of phosphoinositide 3-kinase (PI3K), facilitating its activation by PDK1. mTORC2 may also regulate the actin cytoskeleton through phosphorylation of PRKCA and PXN, and activation of Rho-type guanine nucleotide exchange factors RHOA and RAC1A/RAC1B. mTORC2 also regulates SGK1 phosphorylation (at Ser-422). Additionally, MTOR adjusts CEBPB isoform expression, affecting osteoclastogenesis and plays a significant regulatory role in the circadian clock, influencing the period length and rhythm amplitude of the suprachiasmatic nucleus (SCN) and liver clocks. Finally, MTOR phosphorylates SQSTM1, promoting its interaction with KEAP1 and subsequent inactivation of the KEAP1-CUL3 E3 ubiquitin ligase complex.

Gene References Into Functions

The following publications highlight the diverse roles and regulatory mechanisms of mTOR in various biological processes and disease contexts:

  1. PMID: 28600513
  2. PMID: 22457328
  3. PMID: 29198077
  4. PMID: 28484242
  5. PMID: 30272366
  6. PMID: 29897294
  7. PMID: 30226616
  8. PMID: 30147110
  9. PMID: 30301809
  10. PMID: 30120233
  11. PMID: 30096787
  12. PMID: 28952842
  13. PMID: 29675630
  14. PMID: 29449346
  15. PMID: 29916308
  16. PMID: 29448085
  17. PMID: 29862445
  18. PMID: 28332630
  19. PMID: 30218719
  20. PMID: 30110936
  21. PMID: 29386088
  22. PMID: 29566977
  23. PMID: 30011848
  24. PMID: 28358054
  25. PMID: 30061532
  26. PMID: 30061533
  27. PMID: 29657089
  28. PMID: 30021100
  29. PMID: 29544697
  30. PMID: 29233656
  31. PMID: 30224479
  32. PMID: 29568966
  33. PMID: 29866590
  34. PMID: 29845289
  35. PMID: 29086897
  36. PMID: 29807226
  37. PMID: 29749504
  38. PMID: 29780826
  39. PMID: 29660335
  40. PMID: 29076004
  41. PMID: 29133782
  42. PMID: 29488612
  43. PMID: 27624942
  44. PMID: 29328491
  45. PMID: 30053655
  46. PMID: 28724614
  47. PMID: 29905853
  48. PMID: 29484434
  49. PMID: 29484437
  50. PMID: 29393469

Show All Abstracts Hide Abstracts

Database Links

HGNC: 3942

OMIM: 601231

KEGG: hsa:2475

STRING: 9606.ENSP00000354558

UniGene: Hs.338207

Involvement In Disease
Smith-Kingsmore syndrome (SKS); Focal cortical dysplasia 2 (FCORD2)
Protein Families
PI3/PI4-kinase family
Subcellular Location
Endoplasmic reticulum membrane; Peripheral membrane protein; Cytoplasmic side. Golgi apparatus membrane; Peripheral membrane protein; Cytoplasmic side. Mitochondrion outer membrane; Peripheral membrane protein; Cytoplasmic side. Lysosome. Cytoplasm. Nucleus, PML body. Microsome membrane. Lysosome membrane. Cytoplasmic vesicle, phagosome.
Tissue Specificity
Expressed in numerous tissues, with highest levels in testis.

Q&A

What is mTOR and why is it a significant research target?

mTOR (mechanistic target of rapamycin) is a serine/threonine protein kinase that functions as a central regulator of cellular metabolism, growth, and survival in response to hormones, growth factors, nutrients, energy, and stress signals . It forms two distinct complexes: mTOR Complex 1 (mTORC1) and mTOR Complex 2 (mTORC2). mTORC1 phosphorylates downstream targets like p70S6K and eIF4E to promote mRNA translation and protein synthesis, while mTORC2 affects metabolism and cell survival through AKT phosphorylation and influences the cytoskeleton through PKC alpha activation . Its dysregulation has been implicated in various diseases including cancer, diabetes, and neurological disorders, making it a critical target for biomedical research .

What are the primary applications for mTOR antibodies?

Based on the available research data, mTOR antibodies are primarily used for:

  • Western Blotting (WB) with typical dilutions of 1:1000

  • Immunoprecipitation (IP) with typical dilutions of 1:50

  • Immunofluorescence (IF) for visualization of mTOR localization

  • Simple Western™ assays at dilutions of 1:10-1:50

  • Phosphorylation profiling using antibody arrays to investigate mTOR signaling networks

These applications enable researchers to detect both total mTOR and its phosphorylated forms, providing insights into activation status and signaling dynamics within experimental models.

How do I determine which mTOR antibody is appropriate for my experiment?

When selecting an mTOR antibody, consider:

  • Target specificity: Determine whether you need to detect total mTOR (289 kDa) or specific phosphorylated forms (e.g., p-mTOR at Ser2448)

  • Species reactivity: Verify cross-reactivity with your experimental model (human, mouse, rat, or monkey)

  • Application compatibility: Ensure the antibody is validated for your specific application (WB, IP, IF)

  • Antibody type: Choose between monoclonal (higher specificity) or polyclonal (potentially broader epitope recognition)

  • Validation data: Review the antibody's validation in relevant literature and manufacturer data

For comprehensive phosphorylation profiling, consider array-based approaches with multiple antibodies targeting different mTOR pathway components .

Which detection technique is most sensitive for mTOR pathway analysis?

Research comparing immunohistochemistry (IHC), Western blot (WB), and immunofluorescence (IF) has revealed significant differences in their ability to detect mTOR pathway components. No single technique can effectively detect all mTOR pathway molecules simultaneously . Based on comparative studies:

  • IHC is most sensitive for detecting p-mTOR (Ser2448) and p-p70S6K (Thr389)

  • WB shows superior sensitivity for p70S6K, p-S6Rb, and p-4EBP1 (Thr37/46)

  • IF demonstrates best results for detecting total mTOR (Ser235/236)

  • Only monoclonal p-p70S6k was detectable by two methods (IHC and IF)

This variability highlights the importance of using complementary techniques when comprehensively analyzing the mTOR pathway, especially when correlating results with clinical endpoints such as drug responsiveness.

How should I optimize lysis conditions for mTOR complex detection?

The choice of lysis buffer significantly impacts which mTOR interactions can be detected. Research has shown that detergent selection is particularly critical:

  • Digitonin (1%) has been found to identify the largest number of mTOR protein interactions, making it optimal for protein interaction network analysis

  • For mTOR complex integrity, avoid harsh detergents that may disrupt protein-protein interactions

  • When investigating specific mTOR complexes (mTORC1 vs. mTORC2), consider using CHAPS-containing buffers that preserve complex-specific interactions

  • Include appropriate phosphatase inhibitors when studying phosphorylation states

  • Standardize lysis conditions across experimental replicates to ensure reproducibility

What are the critical controls for validating mTOR antibody specificity?

To ensure reliable results with mTOR antibodies, implement these essential controls:

  • Knockout (KO) or knockdown validation: Demonstrate antibody specificity using mTOR-knockout or mTOR-depleted samples

  • Overexpression systems: Validate antibody performance using cells overexpressing mTOR

  • Blocking peptides: Use competing peptides to confirm signal specificity

  • Phosphorylation-state specificity: For phospho-specific antibodies, treat samples with phosphatases

  • Multiple antibody validation: Screen 3-5 antibodies for each target to identify optimal pairs that can simultaneously bind in the native state

  • Cross-reactivity assessment: Test against closely related kinases in the PI3K/AKT pathway

These validation steps are essential as research has shown that antibody specificity can significantly impact the interpretation of mTOR pathway activation in experimental and clinical samples.

How can I effectively study mTOR protein interaction networks?

For comprehensive analysis of mTOR protein interaction networks (PINs), consider implementing quantitative multiplex immunoprecipitation (QMI):

  • Selection criteria for antibody panels should include:

    • Known relevance to mTOR signaling

    • Established co-associations in protein complexes (verified in databases like BioGrid and IntACT)

    • Association with relevant disease models

    • Availability of suitable, commercially validated antibodies

  • Methodology optimization:

    • Simultaneous immunoprecipitation of multiple protein targets onto flow cytometry beads

    • Detection using fluorescently labeled "probe" antibodies binding to different epitopes

    • 1% digitonin-based lysis conditions to maximize detected interactions

  • Validation approach:

    • Screen multiple antibodies for each target (3-5 recommended)

    • Identify pairs that can simultaneously bind in the native state

    • Validate specificity using knockout or overexpression strategies

This approach enables the detection of dynamic changes in the mTOR PIN under different experimental conditions, revealing coordinated groups of interactions responding to specific signals.

How do I address discrepancies between different detection methods for mTOR pathway components?

When facing contradictory results across detection methods:

  • Acknowledge methodological limitations: Research has conclusively demonstrated that no single molecule in the mTOR pathway is consistently detected by all common techniques (IHC, WB, and IF)

  • Implement a multi-method approach:

    • Use IHC for detecting p-mTOR and p-p70S6K

    • Employ WB for p70S6K, p-S6Rb, and p-4EBP1

    • Apply IF for total mTOR detection

  • Consider epitope accessibility issues:

    • Fixation methods may affect epitope recognition differently across techniques

    • Protein conformation in native versus denatured states influences antibody binding

    • Cross-linking can mask or alter key phosphorylation sites

  • Standardize methods for comparative studies:

    • Establish consistent protocols within research groups

    • Document detailed methodological parameters when publishing

    • Correlate molecular findings with functional outcomes

This approach acknowledges that methodological variations significantly impact the detection of mTOR pathway activation, requiring careful interpretation when correlating with clinical endpoints.

What strategies can improve detection of transient mTOR pathway activation?

Detecting transient mTOR signaling events requires specialized approaches:

  • Temporal sampling design:

    • Implement detailed time-course experiments with closely spaced sampling points

    • Consider rapid fixation methods to "freeze" signaling states

    • Use synchronized cell populations when possible

  • Technical optimization:

    • Employ phosphatase inhibitors immediately upon sample collection

    • Consider in situ detection methods that minimize processing time

    • Optimize antibody concentration and incubation times for maximum sensitivity

  • Signal amplification methods:

    • Utilize tyramide signal amplification for IF applications

    • Consider proximity ligation assays for detecting specific protein interactions

    • Implement phospho-flow cytometry for single-cell resolution of pathway activation

  • Complementary approaches:

    • Correlate antibody-based detection with functional readouts (e.g., downstream target phosphorylation)

    • Use genetic reporters for real-time monitoring when feasible

    • Consider mass spectrometry validation of key phosphorylation events

These strategies help overcome the challenge of capturing dynamic signaling events that might be missed using standard detection protocols.

How can mTOR antibodies be optimized for studying immunological responses?

When investigating mTOR's role in immune signaling pathways:

  • Consider tissue-specific optimization:

    • For marginal zone (MZ) B cells, which mount antibody responses through TACI-dependent pathways, select antibodies that can detect mTOR interactions with MyD88 and TACI

    • Ensure compatibility with lymphoid tissue preparations which may require specialized fixation protocols

  • Functional correlation approach:

    • Pair mTOR detection with measurements of downstream effects like IgG class switching and plasmablast differentiation

    • Correlate mTOR activation with NF-κB signaling, a key transcription factor in the pathway

  • Inhibitor studies:

    • Include rapamycin treatment conditions as controls for mTOR-specific effects

    • Consider monitoring both mTORC1 and mTORC2-specific outputs when studying immune cells

This specialized approach helps delineate how mTOR coordinates immunometabolic reconfiguration in specific lymphocyte populations, particularly in the context of T-cell-independent antibody responses.

What considerations are critical when using mTOR antibodies in cancer research models?

For cancer-focused mTOR pathway investigations:

  • Sample preparation considerations:

    • Optimize protocols for formalin-fixed paraffin-embedded (FFPE) tissue specimens, commonly used in clinical samples

    • Standardize antibody validation using appropriate positive and negative controls from relevant cancer tissues

  • Technique selection based on research questions:

    • For predictive biomarker testing in clinical samples, determine whether IHC, WB, or IF is most appropriate based on the specific mTOR pathway component being investigated

    • Consider the differential sensitivity of techniques for detecting specific phosphorylation sites

  • Cross-validation approach:

    • Implement multiple detection methods for critical findings

    • Correlate protein detection with functional outputs

    • Consider orthogonal validation using genetic approaches

  • Clinical correlation design:

    • When correlating mTOR pathway activation with drug response, standardize methodological approaches

    • Document detailed protocols to enable reproduction across research sites

    • Consider tissue heterogeneity effects on detection sensitivity

These considerations help address the significant methodological challenges in translating mTOR pathway findings from bench research to clinical applications in oncology.

How can I address non-specific binding when using mTOR antibodies?

When experiencing high background or non-specific signals:

  • Antibody optimization:

    • Titrate antibody concentrations (start with manufacturer recommendations like 1:1000 for WB and adjust as needed)

    • Increase washing duration and frequency

    • Try alternative blocking agents (BSA, milk, commercial blockers)

  • Sample preparation modifications:

    • Optimize lysis conditions (consider 1% digitonin which has shown good results for mTOR complex preservation)

    • Pre-clear lysates before immunoprecipitation

    • Reduce sample protein concentration to minimize non-specific interactions

  • Detection system adjustments:

    • For fluorescent detection, include autofluorescence controls

    • With colorimetric methods, optimize substrate development time

    • Consider alternative secondary antibodies from different manufacturers

  • Validation approaches:

    • Include isotype controls at equivalent concentrations

    • Perform competition experiments with blocking peptides

    • Test antibody on samples with known mTOR expression levels

These strategies help distinguish true mTOR signals from background noise, improving data quality and reproducibility.

Why might I observe discrepancies in mTOR molecular weight across different experiments?

Variations in observed molecular weight for mTOR (expected at 289 kDa) may occur due to:

  • Technical factors:

    • Gel percentage and running conditions affect migration patterns

    • Buffer systems can influence apparent molecular weight

    • Incomplete denaturation may result in aberrant migration

    • Prestained markers can show variability between manufacturers

  • Biological considerations:

    • Post-translational modifications alter migration patterns

    • Different mTOR isoforms or splice variants may be detected

    • Proteolytic processing during sample preparation

    • Complex formation that persists through sample preparation

  • Troubleshooting approach:

    • Use high-quality molecular weight standards

    • Include positive control samples with known mTOR expression

    • Consider gradient gels for improved resolution of high molecular weight proteins

    • Optimize denaturation conditions (temperature, time, reducing agents)

Understanding these factors helps researchers interpret unexpected migration patterns and avoid misidentification of mTOR in experimental samples.

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.