LAMTOR5 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism of LAMTOR5 Antibody

LAMTOR5 antibody is a polyclonal or monoclonal immunoglobulin engineered to bind specifically to the LAMTOR5 protein. Its primary function is to enable the detection and quantification of LAMTOR5 via techniques such as immunohistochemistry (IHC), enzyme-linked immunosorbent assay (ELISA), and Western blotting. The antibody’s specificity is attributed to its ability to recognize epitopes on the LAMTOR5 protein, which is part of the Ragulator complex responsible for amino acid sensing and mTORC1 activation .

The antibody’s mechanism involves binding to LAMTOR5, which interacts with vacuolar H⁺-ATPase (v-ATPase) subunits like ATP6V1A to regulate lysosomal acidification and mTORC1 recruitment . This interaction is crucial for maintaining immune homeostasis and cellular metabolism.

Immunohistochemistry (IHC)

LAMTOR5 antibody is widely used in IHC to assess protein expression levels in tissue samples. For example, in head and neck squamous cell carcinoma (HNSCC), IHC revealed overexpression of LAMTOR5 in primary tumors and metastatic lymph nodes, correlating with poor prognosis .

Key Findings from HNSCC Studies:

  • LAMTOR5 Overexpression: Observed in 210 HNSCC cases, with higher expression linked to lymph node metastasis and lymph node grade .

  • Immunosuppressive Correlations: Co-expression with PD-L1, Galectin-9, VISTA, and B7-H4 suggests a role in modulating the tumor microenvironment .

ELISA and Western Blotting

ELISA-based LAMTOR5 antibodies enable precise quantification of protein levels in serum or cellular lysates, while Western blotting is used to validate protein size and post-translational modifications. These methods are critical for studying LAMTOR5 in autoimmune diseases like systemic lupus erythematosus (SLE), where reduced LAMTOR5 levels in peripheral blood mononuclear cells (PBMCs) correlate with disease severity .

Systemic Lupus Erythematosus (SLE)

In SLE patients, reduced LAMTOR5 expression in PBMCs correlates with impaired lysosomal acidification and aberrant mTORC1 activation, leading to autoimmune responses . Myeloid-specific Lamtor5 knockout mice developed SLE-like symptoms, including inflammatory pathology and autoantibody production, underscoring the antibody’s utility in validating these findings .

Head and Neck Squamous Cell Carcinoma (HNSCC)

IHC studies using LAMTOR5 antibody demonstrated its overexpression in HNSCC tissues compared to normal oral mucosa (Fig. 1) . Key correlations include:

MarkerExpression CorrelationClinical Implication
p-Akt Ser473PositiveHyperactivation of PI3K/Akt pathway
p-S6 Ser235/236PositivemTORC1 pathway activation
PD-L1PositiveImmunosuppressive tumor microenvironment

Fig. 1: Representative IHC images showing LAMTOR5 expression in HNSCC vs. normal tissue (adapted from ).

Challenges and Future Directions

While LAMTOR5 antibody has advanced research in oncology and immunology, challenges remain:

  • Cross-reactivity: Limited data on antibody performance across species or isoforms.

  • Standardization: Variable protocols for IHC and ELISA necessitate optimization.

Future studies may focus on developing theranostic antibodies targeting LAMTOR5 for cancer immunotherapy or autoimmune disease management.

Product Specs

Buffer
PBS with 0.02% sodium azide, 50% glycerol, pH 7.3.
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary based on the purchasing method or location. Please consult your local distributors for specific delivery time details.
Synonyms
HBV X interacting protein antibody; HBV X-interacting protein antibody; HBX interacting protein antibody; HBX-interacting protein antibody; hbxip antibody; HBXIP_HUMAN antibody; Hepatitis B virus X interacting protein antibody; Hepatitis B virus X-interacting protein antibody; LAMTOR5 antibody; Late endosomal/lysosomal adaptor MAPK and MTOR activator 5 antibody; MGC71071 antibody; Ragulator complex protein LAMTOR5 antibody; XIP antibody
Target Names
Uniprot No.

Target Background

Function
As a constituent of the Ragulator complex, LAMTOR5 plays a crucial role in amino acid sensing and the activation of mTORC1. mTORC1 is a signaling complex that promotes cell growth in response to growth factors, energy levels, and amino acids. Activated by amino acids through a mechanism involving the lysosomal V-ATPase, the Ragulator acts as a guanine nucleotide exchange factor, activating the small GTPases Rag. Activated Ragulator and Rag GTPases serve as a scaffold, recruiting mTORC1 to lysosomes, where it is subsequently activated. When complexed with BIRC5, LAMTOR5 interferes with apoptosome assembly, preventing the recruitment of pro-caspase-9 to oligomerized APAF1. This interaction selectively suppresses apoptosis initiated via the mitochondrial/cytochrome c pathway. Furthermore, LAMTOR5 down-regulates hepatitis B virus (HBV) replication.
Gene References Into Functions
  1. Research indicates that hepatitis B X-interacting protein (HBXIP) modulates Methyltransferase-like 3 (METTL3) by inhibiting miRNA let-7g, which down-regulates the expression of METTL3 by targeting its 3'UTR. PMID: 29174803
  2. These findings emphasize the potential role of SIRT2 in HBV and HBV-mediated HCC through interaction with HBx. PMID: 29366781
  3. HBXIP can influence the etoposide sensitivity of MCF-7 cell lines. PMID: 29309885
  4. Deacetylation of MST1 mediated by HBXIP-enhanced HDAC6 results in MST1 degradation in a CMA manner, promoting breast cancer growth. PMID: 26657153
  5. High-level expression of HBXIP is associated with the progression of non-small-cell lung cancer and may serve as a valuable biomarker for poor prognostic evaluation and a potential molecular therapy target for patients with non-small-cell lung cancer. PMID: 28718367
  6. HBXIP can suppress gluconeogenesis in hepatoma cells by down-regulating PCK1 to promote hepatocarcinogenesis, involving the miR-135a/FOXO1 axis and the PI3K/Akt/p-FOXO1 pathway. PMID: 27609066
  7. Our research suggests that the oncoprotein HBXIP contributes to abnormal lipid metabolism in breast cancer. PMID: 26980761
  8. HBXIP can act as a mediator protein for DNA damage response signals, activating the G2/M checkpoint to maintain genome integrity and prevent cell death. PMID: 28103177
  9. HBXIP promotes cisplatin resistance and regulates CD147 via Sp1 in ovarian cancer. PMID: 28056551
  10. The mRNA levels of ACSL1 were positively correlated with those of HBXIP in clinical breast cancer tissues. Therefore, we conclude that the oncoprotein HBXIP up-regulates ACSL1 through activating the transcriptional factor Sp1 in breast cancer. PMID: 28132807
  11. Data suggest that HBXIP overexpression appears to be associated with cervical cancer progression and may potentially serve as a biomarker for the early diagnosis, prognostic evaluation, and therapeutic target for cervical cancer. PMID: 28093193
  12. Multivariate analysis indicated that HBXIP, in addition to clinical stage, was a significant independent prognostic factor in patients with ovarian cancer. PMID: 28388957
  13. Expression of HBXIP was significantly increased in UCB tissues. Studies showed that suppression of HBXIP induced cell cycle arrest and increased cell apoptosis in T24 cells. Furthermore, suppression of HBXIP also decreased T24 and PC3 cell proliferation, migration, and invasion. Notably, inhibition of HBXIP reduced tumorigenesis in vivo, suggesting that HBXIP plays a crucial role in disease progression. PMID: 27831760
  14. Upon HBV infection, cellular mechanisms involving SETDB1-mediated H3K9me3 and HP1 induce silencing of HBV cccDNA transcription through modulation of chromatin structure. PMID: 26143443
  15. Therefore, we conclude that the oncoprotein HBXIP up-regulates FGF4 through activating transcriptional factor Sp1 to promote the migration of breast cancer cells. Therapeutically, HBXIP may serve as a novel target in breast cancer. PMID: 26828265
  16. Results support a model where the HBXIP/Hotair/LSD1 complex serves as a critical effector of c-Myc in activating transcription of its target genes, providing insights into how c-Myc drives carcinogenesis. PMID: 26719542
  17. Highly expressed HBXIP accelerates the MDM2-mediated degradation of p53 in breast cancer through modulating the feedback loop of MDM2/p53, resulting in rapid growth of breast cancer cells. PMID: 26229107
  18. Our research suggested that high HBXIP is associated with the progression of breast cancer. HBXIP could be a valuable prognostic marker as well as a potential molecular therapy target for breast cancer patients. PMID: 25178941
  19. HBXIP promotes the migration of breast cancer cells through modulating microtubule acetylation mediated by GCN5. PMID: 25686500
  20. Data show that HBXIP was able to stimulate the activity of Skp2 promoter via transcription factor Sp1, thus promoting the migration of ovarian cancer cells. PMID: 24788380
  21. Hepatitis B protein HBx accelerates hepatocarcinogenesis with partner survivin through modulating tumor suppressor miR-520b and oncoprotein HBXIP. PMID: 24886421
  22. HBXIP facilitates the proliferation of hepatoma cells by up-regulating SCG3 via E2F1 and miR-509-3p modulation. PMID: 24882622
  23. The oncoprotein HBXIP enhances angiogenesis and growth of breast cancer through modulating FGF8 and VEGF. PMID: 24464787
  24. HBXIP nuclear import requires interaction with c-Fos and phosphorylation of both proteins in breast cancer cells. PMID: 23667255
  25. We conclude that the oncoprotein HBXIP, as a co-activator of TF II D, transactivates Lin28B promoter via directly binding to TBP to upregulate the expression of Lin28B, promoting the proliferation of breast cancer cells. PMID: 23494474
  26. Knockdown of HBXIP rescued the inhibition of HBV that occurred after the loss of miR-501 in HepG2.2.15 cells, suggesting that miR-501 induced HBV replication partially by targeting HBXIP. PMID: 23266610
  27. HBXIP promotes the proliferation of breast cancer cells via upregulating PDGFB. PMID: 23537647
  28. The oncoprotein HBXIP is able to activate the transcriptional coregulatory protein LMO4 through transcription factor Sp1, promoting the proliferation of breast cancer cells. PMID: 23291272
  29. It was found that HBXIP was able to stimulate the promoter of Skp2 through binding to the -640/-443 region in Skp2 promoter involving activating E2F transcription factor 1. PMID: 23352642
  30. This study identified HBXIP and C7orf59 as two additional Ragulator components that are required for mTORC1 activation by amino acids. PMID: 22980980
  31. HBXIP up-regulates S100A4 through activating S100A4 promoter involving STAT4 and inducing PTEN/PI3K/AKT signaling to promote growth and migration of breast cancer cells. PMID: 22740693
  32. Overexpression of HBXIP increased HepG2 cell-induced endothelial cells migration, proliferation, and angiogenesis, which may be related to increasing phosphorylation of endothelial NO synthase in HUVECs. PMID: 22209835
  33. Data suggest that HBXIP upregulates CD46, CD55, and CD59 through p-ERK1/2/NF-kappaB signaling to protect breast cancer from complement-dependent cytotoxicity. PMID: 22293503
  34. The different structure forms of HBx protein influence their intracellular distribution in hepatocellular carcinoma HepG2 cells. PMID: 21651858
  35. miR-520b is involved in regulating breast cancer cell migration by targeting HBXIP and IL-8 via a network in which HBXIP promotes migration by stimulating NF-kappaB-mediated IL-8 expression. PMID: 21343296
  36. The x gene of HBV (HBx) is the most common open reading frame integrated into the host genome in hepatocellular carcinoma, and the integrated HBx is frequently mutated in hepatocellular carcinoma. PMID: 20811532
  37. Hepatitis B virus pX interacts with HBXAP. PMID: 11788598
  38. Elevated levels of GDN/PN1 and XIP mRNAs induced by Allitridi provide valuable molecular evidence for elucidating garlic's efficacies against neurodegenerative and inflammatory diseases. PMID: 11925594
  39. Survivin-HBXIP complexes, but neither survivin nor HBXIP individually, bind pro-caspase-9, preventing its recruitment to Apaf1, and thereby selectively suppressing apoptosis. PMID: 12773388
  40. HBXIP up-regulates LTA expression in hepatocytes. PMID: 15955450
  41. HBXIP sensitizes HepG2 cells to UV light-induced DNA damage. PMID: 16055925
  42. Suppressor of var1, 3-like 1 protein interacts with HBXIP, previously identified as a cofactor of survivin in suppression of apoptosis. PMID: 16176273
  43. The codon-38 change in genotype C is an independent risk factor for the development of HCC and may serve as a useful molecular marker for predicting clinical outcomes in patients infected with HBV. PMID: 17050029
  44. One of the functions of HBXIP is its involvement in cell proliferation. PMID: 17303008
  45. Hepatitis B virus X protein stimulates the mitochondrial translocation of Raf-1 via oxidative stress. PMID: 17428866
  46. HBXIP is a critical target of viral HBx for promoting genetic instability through the formation of defective spindles and subsequent aberrant chromosome segregation. PMID: 18032378
  47. HBXIP significantly stimulated the transcription and expression of telomerase reverse transcriptase and increased the activity of telomerase. PMID: 18158869
  48. The overexpression of survivin in the majority of NSCLCs, together with the abundant or upregulated expression of HBXIP and XIAP, suggests that tumors are endowed with resistance against a variety of apoptosis-inducing conditions. PMID: 19885569

Show More

Hide All

Database Links

HGNC: 17955

OMIM: 608521

KEGG: hsa:10542

STRING: 9606.ENSP00000256644

UniGene: Hs.439815

Protein Families
LAMTOR5 family
Subcellular Location
Lysosome. Cytoplasm, cytosol.
Tissue Specificity
Highly expressed in skeletal and cardiac muscle, followed by pancreas, kidney, liver, brain, placenta and lung. Elevated levels in both cancerous and non-cancerous liver tissue of patients with chronic HBV infection compared with hepatic tissue without HB

Q&A

What is LAMTOR5 and what cellular functions does it regulate?

LAMTOR5 is a component of the Ragulator complex that plays essential roles in several cellular functions. It serves as a late endosomal/lysosomal adaptor and activator for both MAPK and mTOR signaling pathways . LAMTOR5 physically associates with ATP6V1A, an essential subunit of vacuolar H⁺-ATPase (v-ATPase), and promotes the V0/V1 holoenzyme assembly to facilitate lysosome acidification . This protein is critical for immune homeostasis by integrating v-ATPase activity, lysosome function, and mTOR pathway regulation. Recent research has established that LAMTOR5 defects can lead to autoimmune conditions resembling systemic lupus erythematosus (SLE) . Additionally, LAMTOR5 has been identified as a key regulator in TLR4 signaling and inflammation control, as it associates with TLR4 and facilitates their colocalization at autolysosomes, affecting inflammatory response magnitudes .

What alternative names exist for LAMTOR5 in the scientific literature?

LAMTOR5 is known by several synonyms in scientific literature, which is important to recognize when conducting comprehensive literature searches:

  • HBV X-interacting protein (HBXIP)

  • HBX-interacting protein

  • Hepatitis B virus X-interacting protein

  • Ragulator complex protein LAMTOR5

  • XIP

Understanding these alternative nomenclatures ensures thorough literature review and prevents researchers from missing relevant publications when investigating this protein.

What are the optimal applications for anti-LAMTOR5 antibodies in research?

Based on available data, anti-LAMTOR5 antibodies have been successfully validated for immunohistochemistry (IHC) applications . The antibody detects endogenous levels of total LAMTOR5 protein and shows specificity for human samples . While IHC is a well-validated application, researchers should consider that different antibodies may have varying specificities and optimal applications. When designing experiments, it's important to select antibodies that have been validated for your specific application and sample type. For optimal results in IHC applications, the recommended dilution is 1/100, as demonstrated in validation studies with human tonsil and thyroid cancer tissue samples .

What methodologies can be used to study LAMTOR5's role in lysosomal acidification?

To investigate LAMTOR5's role in lysosomal acidification, researchers can employ several complementary approaches:

  • LysoSensor assays: LysoSensor Yellow/Blue DND-160, a dual-emission ratiometric probe, can be used to measure lysosomal pH in LAMTOR5 wildtype versus knockout/knockdown cells .

  • Co-immunoprecipitation studies: These can detect physical interaction between LAMTOR5 and ATP6V1A (v-ATPase subunit) to understand the molecular basis for LAMTOR5's effect on acidification .

  • Confocal microscopy: This technique allows visualization of lysosomal markers and acidification in live cells under different conditions.

  • Autophagic flux assessment: Using tandem mCherry-EGFP-LC3B reporter systems can help visualize acidified autophagic structures, which appear as red puncta (since GFP is more sensitive to acidic conditions while mCherry remains stable) .

  • Biochemical assays: Measuring the ratio of LC3-II/I and p62 levels can indicate autophagosome formation and maturation, which are affected by lysosomal acidification and function .

What controls should be included when using LAMTOR5 antibodies for immunohistochemistry?

When using LAMTOR5 antibodies for immunohistochemistry, several controls should be included to ensure specificity and validity of results:

  • Negative controls:

    • Omission of primary antibody

    • Isotype control (non-specific IgG from the same species)

    • Tissues known not to express LAMTOR5

  • Positive controls:

    • Human tonsil tissue, which has been validated to show LAMTOR5 expression

    • Human thyroid cancer tissue, which also demonstrates LAMTOR5 expression

  • Blocking controls:

    • Pre-incubation with fusion protein (as shown in validation data where fusion protein treatment blocks the antibody signal)

  • Dilution optimization:

    • Test different antibody dilutions (starting with manufacturer's recommendation of 1/100)

  • Secondary antibody controls:

    • Suitable secondary antibodies include: Goat Anti-Rabbit IgG H&L with various conjugates (AP, Biotin, FITC, or HRP) depending on detection method

How does LAMTOR5 deficiency impact macrophage phenotype and function?

LAMTOR5 deficiency profoundly alters macrophage phenotype and function, shifting them toward a hyperactivated, inflammatory state. Experimental data shows that LAMTOR5 knockout macrophages exhibit:

  • Upregulated surface markers: Increased expression of MHCII and costimulatory molecules (CD80, CD86, and CD40)

  • Enhanced phagocytic function: Significantly increased phagocytic capacity compared to control cells

  • Altered cytokine profile:

    • Increased production of proinflammatory cytokines and M1 marker genes

    • Repressed expression of immunosuppressive cytokines and M2-featured genes

  • Dysregulated gene expression: Differential expression of 42 lysosome-associated genes, including Rilpl2, Atp6v0d2, TMEM150a, and IFITM3

  • Impaired lysosomal acidification: Elevated lysosomal pH compared to wildtype macrophages

  • Altered autophagy: Reduced LC3-II/I ratio (indicating impaired autophagosome formation) and elevated p62 levels (suggesting decreased autophagic degradation)

This shift toward an inflammatory phenotype in LAMTOR5-deficient macrophages contributes to the development of autoimmune-like conditions observed in experimental models and potentially in human disease conditions like SLE.

What is the relationship between LAMTOR5 and TLR4 signaling in inflammatory responses?

LAMTOR5 plays a crucial regulatory role in TLR4 signaling and inflammatory response control through several mechanisms:

  • Direct TLR4 interaction: LAMTOR5 associates with TLR4 via their LZ/TIR domains, forming a physical complex

  • Subcellular colocalization: LAMTOR5 facilitates the colocalization of TLR4 at autolysosomes following LPS stimulation

  • mTORC1 regulation: LAMTOR5 prevents lysosomal tethering and activation of mTORC1 upon LPS stimulation

  • TFEB derepression: By modulating mTORC1 activity, LAMTOR5 derepresses TFEB (Transcription Factor EB), promoting autophagic degradation of TLR4

  • Inflammatory resolution: LAMTOR5 contributes to the timely degradation of TLR4, helping to resolve inflammatory responses after pathogen recognition

In LAMTOR5-deficient conditions, TLR4 degradation is delayed, leading to sustained inflammatory signaling and increased susceptibility to endotoxic shock. Notably, Lamtor5 haploinsufficient mice showed increased mortality during endotoxic shock, demonstrating the physiological importance of this regulatory pathway .

How does LAMTOR5 integrate nutrient sensing with immune regulation?

LAMTOR5 serves as a critical molecular bridge between metabolic sensing and immune regulation through several interconnected mechanisms:

  • Nutrient-dependent immune modulation: Nutrient deprivation, particularly leucine deprivation, blunts inflammatory signaling and provides protection during endotoxic shock, an effect that is largely abrogated upon LAMTOR5 deletion

  • Amino acid sensing machinery component: As part of the Ragulator complex, LAMTOR5 contributes to amino acid sensing and subsequent mTORC1 activation or inhibition

  • GEF function for RAG GTPase: LAMTOR5, along with other Lamtor proteins, functions as a guanine exchange factor for RAG GTPase to transduce nutrient-sensing signals

  • Autolysosomal pathway regulation: Through modulation of TFEB activity, LAMTOR5 influences autophagy processes that are essential for both metabolic adaptation and immune response resolution

This integration of pathogenic signals and nutrient availability through LAMTOR5 represents a homeostatic mechanism that optimizes inflammatory responses based on metabolic status. This function has significant implications for understanding and potentially treating TLR4-associated inflammatory and metabolic disorders .

What are common challenges when working with LAMTOR5 antibodies and how can they be addressed?

When working with LAMTOR5 antibodies, researchers may encounter several challenges:

  • Cross-reactivity concerns:

    • Solution: Validate antibody specificity using positive and negative controls, including fusion protein blocking experiments as demonstrated in validation studies

    • Approach: Compare results using multiple antibodies targeting different epitopes of LAMTOR5

  • Variable expression levels:

    • Solution: Optimize sample preparation and antibody concentration

    • Approach: For IHC applications, the recommended starting dilution is 1/100, but titration may be necessary for specific tissues

  • Storage and stability issues:

    • Solution: Store antibody at -20°C as recommended

    • Approach: Avoid repeated freeze-thaw cycles and prepare working aliquots

  • Detection sensitivity:

    • Solution: Choose appropriate secondary antibodies based on application

    • Approach: For LAMTOR5 rabbit polyclonal antibodies, suitable secondaries include Goat Anti-Rabbit IgG H&L with various conjugates (AP, Biotin, FITC, or HRP)

  • Background signal:

    • Solution: Optimize blocking conditions and washing steps

    • Approach: Include appropriate negative controls in each experiment

How can researchers effectively study the relationship between LAMTOR5 and lysosomal function?

To effectively study the relationship between LAMTOR5 and lysosomal function, researchers should consider a multi-faceted approach:

  • Genetic manipulation:

    • Generate LAMTOR5 knockout/knockdown models using CRISPR-Cas9 or shRNA

    • Create cell-specific conditional knockout models (e.g., myeloid-specific Lamtor5 knockout mice as described in the literature)

  • Lysosomal pH measurement:

    • Use LysoSensor Yellow/Blue DND-160 for dual-emission ratiometric pH measurement

    • Compare pH in LAMTOR5-sufficient and LAMTOR5-deficient cells

  • Protein interaction studies:

    • Perform co-immunoprecipitation to detect LAMTOR5 interaction with ATP6V1A and other v-ATPase components

    • Use proximity ligation assays to visualize protein interactions in situ

  • Autophagy assessment:

    • Monitor LC3-II/I ratios and p62 levels by Western blotting

    • Use mCherry-EGFP-LC3B reporter systems to visualize autophagic flux

  • Transcriptional profiling:

    • Conduct RNA-seq to identify differentially expressed lysosome-associated genes

    • Perform GO analysis to identify enriched pathways related to lysosomal function

  • Functional assays:

    • Assess lysosomal enzyme activity

    • Measure degradation rates of known lysosomal substrates

What are promising therapeutic applications based on LAMTOR5 biology?

Based on current understanding of LAMTOR5 biology, several promising therapeutic applications could be developed:

  • Autoimmune disease treatment: Since LAMTOR5 deficiency leads to SLE-like autoimmunity, strategies to restore or enhance LAMTOR5 function could potentially treat autoimmune conditions. The research indicates that enforced expression of LAMTOR5 resumed lysosomal acidity in PBMCs from SLE patients .

  • Anti-inflammatory approaches: Targeting the LAMTOR5-TLR4 interaction could provide novel ways to modulate inflammatory responses in conditions like sepsis. This is supported by findings that LAMTOR5 haploinsufficient mice showed increased mortality during endotoxic shock .

  • Metabolic inflammation targeting: The unique position of LAMTOR5 at the intersection of nutrient sensing and immune regulation makes it a potential target for treating metabolic disorders with inflammatory components. The finding that leucine deprivation blunts inflammatory signaling through LAMTOR5-dependent mechanisms suggests nutritional or pharmacological approaches .

  • Cancer therapy: Given LAMTOR5's associations with oncogenic proteins like c-FOS, c-Myc, Survivin, and p53, as well as its role in promoting proliferation in cancer cells, targeting LAMTOR5 might represent a novel approach in cancer treatment .

  • Lysosomal storage disease applications: LAMTOR5's role in lysosomal acidification suggests potential applications in lysosomal storage diseases, where lysosomal function is compromised.

What are key unresolved questions regarding LAMTOR5 function?

Despite recent advances, several key questions about LAMTOR5 remain unresolved:

  • Tissue-specific functions: How does LAMTOR5 function differ across various tissue and cell types beyond macrophages and immune cells?

  • Regulatory mechanisms: What regulates LAMTOR5 expression and activity under different physiological and pathological conditions?

  • Structural biology: What are the precise structural determinants of LAMTOR5's interactions with TLR4, v-ATPase, and Rag GTPase?

  • Systems biology perspective: How does LAMTOR5 coordinate its multiple functions in nutrient sensing, immune regulation, and lysosomal function in an integrated cellular context?

  • Evolutionary significance: How conserved is LAMTOR5 function across species, and what does this reveal about the co-evolution of metabolic and immune systems?

  • Disease relevance beyond autoimmunity: What roles might LAMTOR5 play in neurodegeneration, metabolic diseases, or other conditions where lysosomal function is implicated?

  • Interaction with other Ragulator components: How do the different components of the Ragulator complex (Lamtor1-5) coordinate their functions, and are there redundancies?

What methodological advances might improve LAMTOR5 research?

Future advances in LAMTOR5 research could benefit from several methodological improvements:

  • Live-cell imaging techniques: Development of specific fluorescent tags for LAMTOR5 that maintain protein function would allow real-time visualization of its dynamics and interactions.

  • Tissue-specific conditional knockout models: Generation of additional tissue-specific LAMTOR5 knockout models would help delineate its function in different physiological contexts.

  • High-throughput screening approaches: Development of assays suitable for identifying small molecule modulators of LAMTOR5 function could accelerate therapeutic development.

  • Improved antibodies and detection methods: Development of highly specific monoclonal antibodies against different epitopes of LAMTOR5 would enhance research capabilities.

  • Proteomics approaches: Comprehensive interactome studies would help identify the full range of LAMTOR5 binding partners across different cellular conditions.

  • Single-cell analysis: Application of single-cell technologies could reveal cell-to-cell variation in LAMTOR5 function and its consequences.

  • Structural biology techniques: Cryo-EM or X-ray crystallography studies of LAMTOR5 in complex with its binding partners would provide mechanistic insights at the atomic level.

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.