MITF Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery time may vary based on your purchase method and location. For specific delivery timelines, please consult your local distributors.
Synonyms
BHLHE32 antibody; bHLHe32 antibody; Class E basic helix-loop-helix protein 32 antibody; CMM8 antibody; Homolog of mouse microphthalmia antibody; Mi antibody; Microphthalmia associated transcription factor antibody; Microphthalmia, mouse, homolog of antibody; Microphthalmia-associated transcription factor antibody; MITF antibody; MITF_HUMAN antibody; mitfa antibody; nacre antibody; WS2 antibody; WS2A antibody; z3A.1 antibody
Target Names
Uniprot No.

Target Background

Function
MITF is a transcription factor that plays a crucial role in regulating the expression of genes involved in cell differentiation, proliferation, and survival. It binds to specific DNA sequences known as M-boxes (5'-TCATGTG-3') and E-boxes (5'-CACGTG-3'), located in the promoters of target genes such as BCL2 and tyrosinase (TYR). MITF is particularly important in melanocyte development, where it controls the expression of tyrosinase (TYR) and tyrosinase-related protein 1 (TYRP1). Furthermore, MITF plays a critical role in the differentiation of various cell types, including neural crest-derived melanocytes, mast cells, osteoclasts, and optic cup-derived retinal pigment epithelium.
Gene References Into Functions
  1. Studies have shown that MITF is highly expressed in myeloma cells and regulates cdk2 expression, contributing to cell resistance to BRAF and Hsp90 inhibitors. PMID: 29507054
  2. Research suggests that glycogen synthase kinase 3 (GSK3) and BRAF/MAPK signaling pathways converge to regulate the nuclear export of MITF. PMID: 30150413
  3. Our findings provide new insights into how mutations in MITF can lead to different phenotypes of Waardenburg syndrome type 2 through the Wnt/beta-catenin signaling pathway. PMID: 29531335
  4. This study reports a novel mutation (c.718C>G; p. (Arg240Gly)) in the MITF gene, associated with hearing loss in Han people. PMID: 29484430
  5. This research investigated the upregulation of microphthalmia-associated transcription factor (Mitf) and enhanced melanogenesis induced by Cymbopogon schoenanthus phenol extracts. PMID: 29359158
  6. The essential melanocyte-specific transcription factor MITF regulates the expression of the MYO5A gene, which encodes the molecular motor myosin-Va. PMID: 27939378
  7. Observations suggest that primary and metastatic melanomas comprise not only MITF-high and MITF-low cells but also subpopulations expressing markers of both signatures. These diverse cell populations may be intermingled, contributing to the spatial heterogeneity of tumors. PMID: 28855355
  8. SH3BP4 is transcriptionally regulated by MITF as a direct target. PMID: 28819321
  9. This study demonstrates that the FANC pathway operates downstream of MiTF, establishing an epistatic relationship between MiTF and the FANC pathway. PMID: 27827420
  10. MITF expression levels in hepatic cancer cells may be determined by the balance between Hedgehog signaling and cellular stress. PMID: 28794318
  11. Data strongly suggest that glucose deprivation suppresses MITF expression through reactive oxygen species-induced ATF4 up-regulation, leading to reduced melanoma cell proliferation. PMID: 28380427
  12. Overexpression of MITF-A resulted in a significant increase in nephron number and larger kidneys, while Mitfa deficiency led to reduced nephron number. PMID: 29240767
  13. MITF may play a role in the development of acquired drug resistance through hyper-activation of the PI3K pathway. PMID: 27391157
  14. Mutations in the MITF gene are associated with Waardenburg syndrome type 2A. PMID: 29094203
  15. A sumoylation-defective germline mutation in MITF, a master regulator of melanocyte homeostasis, is linked to melanoma development. [review] PMID: 28825724
  16. Single Nucleotide Polymorphism in the MITF gene is associated with facial solar lentigines. PMID: 27327535
  17. Phosphorylation of MITF by AKT influences its downstream targets and causes TP53-dependent cell senescence. PMID: 27702651
  18. This study identified two novel MITF mutations in patients with Tietz/Waardenburg syndrome type 2A, suggesting that posterior microphthalmos might be a clinical characteristic of this syndrome. PMID: 27604145
  19. Data show that poly(ADP-ribose) polymerase 1 (PARP1)-mediated senescence rescue was accompanied by transcriptional activation of MITF, suggesting a role for PARP1 in melanomagenesis. PMID: 28759004
  20. MITF is a direct target of miR-137. PMID: 26845432
  21. In melanoma cell lines, ILEI is highly expressed in MITF-low invasive cells, and switching between MITF-low invasive and MITF-high proliferative states can alter ILEI expression. PMID: 28545079
  22. Suppression of MITF activity by UCHL1 through protein degradation could potentially lead to new therapeutic approaches for melanoma or dyspigmentation disorders. PMID: 28392346
  23. This study provides novel insights into the effect of Bcl-2 overexpression in melanoma cells, demonstrating that Bcl-2 modulates MITF nuclear activity. PMID: 26599548
  24. This research explores molecular interactions between CRD-BP and MITF mRNA. PMID: 28182633
  25. This study uncovers novel mechanisms linking MITF-dependent inhibition of invasion to suppression of guanylate metabolism. PMID: 27181209
  26. This study demonstrates that androgen receptor (AR) can promote melanoma metastasis by altering the miRNA-539-3p/USP13/MITF/AXL signaling pathway, suggesting that targeting this pathway with AR degradation enhancer ASC-J9 might suppress melanoma metastasis. PMID: 27869170
  27. This review discusses the fundamental roles of MITF in melanocytes and melanoma. PMID: 28263292
  28. This study shows that the MITF-E318K variant reduces the program of senescence, potentially favoring melanoma progression in vivo. PMID: 28376192
  29. The germline variant MITF, p.E318K, is associated with an increased risk of other neural crest-derived tumors such as paraganglioma (PGL). PMID: 27680874
  30. Microenvironmental cues, including inflammation-mediated resistance to adoptive T-cell immunotherapy, can transcriptionally repress MITF via ATF4 in response to inhibition of translation initiation factor eIF2B. PMID: 28096186
  31. GPER enhances melanogenesis through PKA by upregulating microphthalmia-related transcription factor-tyrosinase in melanoma. PMID: 27378491
  32. Data show that TFAP2A binds to many of the same regulatory elements as MITF in melanocytes. PMID: 28249010
  33. This study describes a syndrome, termed COMMAD, characterized by coloboma, osteopetrosis, microphthalmia, macrocephaly, albinism, and deafness. COMMAD is associated with biallelic MITF mutant alleles, suggesting a role for MITF in regulating processes beyond melanocyte development, such as optic-fissure closure and bone development or homeostasis. PMID: 27889061
  34. Data indicate that NFIB protein increases EZH2 protein expression downstream of BRN2 protein, which further decreases MITF protein levels. PMID: 28119061
  35. Overall, in melanoma lymph node metastases, MITF protein expression was not tightly correlated with its gene targets. PMID: 27515936
  36. DRD4 antagonist exhibits an antimelanogenic effect associated with downregulation of MITF transcription through ERK activation. PMID: 26782007
  37. AKT modulates the nuclear translocation of MITF. PMID: 28165011
  38. Studies have established that the cooperative antiproliferative effects of aspirin and I3C in human melanoma cells trigger a significant downregulation of MITF-M gene expression and disruption of MITF-M promoter activity. These results demonstrate that aspirin-regulated Wnt signaling and I3C-targeted signaling pathways converge at distinct DNA elements in the MITF-M promoter to cooperatively disrupt MITF-M expression. PMID: 27055402
  39. Adding MITF>/=50% to the logistic regression analysis significantly improves the accuracy of the melanoma nomogram in predicting regional nodal spread. PMID: 27919990
  40. The MITF p.E318K mutation does not appear to play a major role in sporadic renal cell carcinoma carcinogenesis but may be restricted to a rare subpopulation of inherited renal cell carcinoma. PMID: 26999813
  41. Overexpression of MITF is associated with melanoma cell survival and progression. PMID: 27185926
  42. This research concludes that the expression of Rlbp1 and Rdh5 critically depends on functional Mitf in the retinal pigment epithelium (RPE), suggesting that MITF plays an important role in controlling retinoid processing in the RPE. PMID: 26876013
  43. Concomitant AURKA/BRAF and AURKA/MEK targeting overcame MAPK signaling activation-associated resistance signatures in BRAF- and NRAS-mutated melanomas, respectively, resulting in enhanced anti-proliferative activity and apoptotic cell death. PMID: 26962685
  44. Results show that Mitf, likely including Mitf-M, is expressed in the mitral cells and tufted cells that transmit olfactory sensory neuron information to the olfactory cortex. PMID: 26522736
  45. SOX5 has a strong inhibitory effect on MITF expression and appears to have a significant clinical impact on melanoma during tumor progression. PMID: 26927636
  46. In addition to melanoma risk, MITF p.E318K is associated with a high nevi count and could play a role in fast-growing melanomas. PMID: 26650189
  47. The expression of the molecular marker Mitf in primary cutaneous melanomas is a useful tool for assessing lymph node status. PMID: 26317170
  48. This study suggests an MITF-CEACAM1 axis as a potential determinant of melanoma progression. PMID: 26301891
  49. LEF-1 and MITF regulate tyrosinase gene transcription in vitro by binding to its promoter. PMID: 26580798
  50. This research shows that MITF-A mRNA is predominantly expressed in all three human liver cancer cell lines examined. PMID: 26773496

Show More

Hide All

Database Links

HGNC: 7105

OMIM: 103470

KEGG: hsa:4286

STRING: 9606.ENSP00000295600

UniGene: Hs.166017

Involvement In Disease
Waardenburg syndrome 2A (WS2A); Waardenburg syndrome 2, with ocular albinism, autosomal recessive (WS2-OA); Tietz albinism-deafness syndrome (TADS); Melanoma, cutaneous malignant 8 (CMM8); Coloboma, osteopetrosis, microphthalmia, macrocephaly, albinism, and deafness (COMMAD)
Protein Families
MiT/TFE family
Subcellular Location
Nucleus. Cytoplasm.
Tissue Specificity
Expressed in melanocytes (at protein level).; [Isoform A2]: Expressed in the retinal pigment epithelium, brain, and placenta. Expressed in the kidney.; [Isoform C2]: Expressed in the kidney and retinal pigment epithelium.; [Isoform H1]: Expressed in the k

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : WB

Sample type: cells

Review: The rabbit polyclonal MITF antibody, rabbit polyclonal

Q&A

What is MITF and why is it an important research target?

MITF (Microphthalmia-associated Transcription Factor) is a basic helix-loop-helix-leucine-zipper (bHLH-Zip) transcription factor that plays a crucial role in the development and survival of melanocytes and retinal pigment epithelium . It regulates the transcription of pigmentation enzyme genes including tyrosinase, TRP1, and TRP2 . MITF is particularly important in research because mutations in the MITF gene are associated with several disorders, most notably Waardenburg syndrome type 2A, an auditory-pigmentary syndrome characterized by developmental defects in cells derived from neural crest . Additionally, MITF has emerged as a central player in melanoma biology, making it an essential target for cancer research . The transcription factor functions downstream of oncogenic pathways and microenvironment stimuli that can restrain immune responses, establishing it as a potential melanoma immunotherapy response factor .

What are the different MITF isoforms and how do they impact antibody selection?

At least 12 isoforms of MITF have been identified, which exhibit differential patterns of expression among cell and tissue types . The most well-characterized isoforms include MITF-A, MITF-B, MITF-C, MITF-H, and MITF-M . The MITF-M isoform is particularly important as it is restricted to the melanocyte cell lineage . When selecting an MITF antibody, researchers must consider which isoforms they need to detect based on their experimental model and research question . Some antibodies are raised against N-terminal fragments that may recognize specific isoforms, while others target conserved regions that detect multiple isoforms . The antibody documentation should be carefully reviewed to ensure compatibility with the isoform of interest, especially considering that different isoforms may exhibit varying molecular weights (ranging from 50-75 kDa) .

How do I determine the appropriate MITF antibody species reactivity for my research?

MITF is highly conserved across species, but there are important differences in expression patterns and function that must be considered when selecting an antibody . Available MITF antibodies show reactivity with different species, with many recognizing human, mouse, and rat MITF . When selecting an antibody, verify the validated species reactivity in the product information . For example, the MITF (D5G7V) Rabbit mAb shows reactivity with human, mouse, rat, hamster, and monkey samples , while the polyclonal antibody 13092-1-AP has been tested specifically with human, mouse, and rat samples . If working with less common research models, additional validation may be necessary to confirm cross-reactivity. Species-specific differences in MITF may affect epitope recognition, so preliminary testing with appropriate positive and negative controls is recommended when applying an antibody to a species not explicitly listed in the validation data.

What are the optimal conditions for using MITF antibodies in Western Blotting?

For optimal Western Blotting results with MITF antibodies, consider the following methodological details: First, select the appropriate dilution ratio based on the specific antibody - for example, the Cell Signaling MITF (D5G7V) Rabbit mAb requires a 1:1000 dilution , while the Proteintech polyclonal antibody (13092-1-AP) can be used at dilutions ranging from 1:500 to 1:3000 . Second, be aware of the expected molecular weight range for MITF, which typically appears between 50-75 kDa depending on the isoform and potential post-translational modifications . Observed molecular weights can vary from 59-65 kDa for some antibodies . Third, include appropriate positive controls; validated cell lines include A549 cells, Jurkat cells, NIH/3T3 cells, or tissue samples such as mouse heart tissue or rat skin tissue . Finally, ensure your detection system is sensitive enough, as MITF expression can vary significantly across different cell types and experimental conditions . If you encounter non-specific bands, optimization of blocking conditions or further antibody dilution may be necessary.

How should MITF antibodies be used in ChIP and ChIP-seq experiments?

When using MITF antibodies for Chromatin Immunoprecipitation (ChIP) and ChIP-seq applications, several methodological considerations are crucial for successful experiments. For optimal results with the Cell Signaling MITF (D5G7V) Rabbit mAb, use 10 μl of antibody and 10 μg of chromatin (approximately 4 x 10^6 cells) per immunoprecipitation, with a recommended dilution of 1:50 . This antibody has been validated using SimpleChIP Enzymatic Chromatin IP Kits . For quality control, include appropriate qPCR primers to verify enrichment of known MITF target regions. The Active Motif antibody documentation notes that their Human Negative Control Primer Set 2 has been validated for performing qPCR when using their antibody for ChIP on human samples, while Mouse Positive Control Primer Set Pax-2 and Mouse Negative Control Primer Set 3 are recommended for mouse samples . These controls are essential for distinguishing specific binding from background signal. When moving to ChIP-seq applications, ensure high-quality DNA library preparation and sufficient sequencing depth to capture the full spectrum of MITF binding sites across the genome.

What protocol should be followed for immunofluorescence studies using MITF antibodies?

For immunofluorescence (IF) studies using MITF antibodies, follow this methodological approach to ensure optimal staining and specificity: Begin with appropriate fixation, typically using 4% paraformaldehyde for 10-15 minutes at room temperature, although specific protocols may vary based on the cell type and antibody used . Permeabilization with 0.1-0.5% Triton X-100 is generally suitable for accessing nuclear MITF. When blocking, use 5-10% normal serum (from the species in which the secondary antibody was raised) to minimize background. Antibody dilutions must be optimized; for example, the polyclonal antibody from Proteintech has been successfully used in multiple immunofluorescence studies, though optimal dilutions should be determined empirically for each application . MITF localizes predominantly to the nucleus in melanocytes and melanoma cells, so expect a nuclear staining pattern . Include appropriate positive controls such as melanoma cell lines with known MITF expression and negative controls omitting the primary antibody or using cell types that don't express MITF. To visualize MITF alongside other markers, consider co-staining protocols with attention to preventing antibody cross-reactivity, especially when studying MITF's relationship with the immune system as described in recent research .

Why might MITF detection show variable results across different experimental conditions?

MITF detection variability across experiments can stem from multiple factors that researchers should systematically address. First, the presence of multiple MITF isoforms (at least 12 have been identified) with different expression patterns across tissues and cell types can lead to inconsistent results if antibodies have varying affinities for specific isoforms . Second, post-translational modifications significantly impact MITF function and detection - MITF is known to be phosphorylated by MAP kinase in response to c-kit activation, which upregulates its transcriptional activity but may affect epitope recognition by certain antibodies . Third, fixation and extraction methods can dramatically influence MITF detection, particularly for immunostaining techniques, as improper fixation may mask epitopes or create artifacts . Fourth, MITF expression levels fluctuate naturally in response to microenvironmental conditions, especially in melanoma cells where MITF is a key determinant of cellular plasticity and tumor heterogeneity . Finally, buffer composition can impact antibody performance - for example, the Active Motif MITF antibody is provided in a specific buffer (70 mM Tris pH 8, 105 mM NaCl, 31 mM glycine, 0.07 mM EDTA, 30% glycerol, and 0.035% sodium azide) that maintains optimal antibody activity . When troubleshooting, researchers should systematically modify each of these variables while maintaining appropriate positive and negative controls.

What controls should be included when validating MITF antibody specificity?

To rigorously validate MITF antibody specificity, incorporate these essential controls in your experimental design: First, include positive control samples with well-characterized MITF expression, such as melanoma cell lines for the MITF-M isoform or validated tissue samples like mouse heart tissue and rat skin tissue as recommended for certain antibodies . Second, implement negative controls using cell lines or tissues known not to express MITF, or cells where MITF has been knocked down using siRNA or CRISPR-Cas9 approaches. Third, perform isotype controls using non-specific IgG from the same species as your primary MITF antibody at equivalent concentrations to assess non-specific binding. Fourth, conduct peptide competition assays where the antibody is pre-incubated with the immunizing peptide before application to samples - specific binding should be blocked. Fifth, compare results across multiple MITF antibodies targeting different epitopes, as consistent staining patterns increase confidence in specificity. Finally, correlate protein detection with mRNA expression data using qPCR or RNA-seq approaches. For ChIP applications specifically, include both positive control primer sets targeting known MITF binding regions and negative control primer sets for genomic regions not bound by MITF, such as those validated for the Active Motif MITF antibody .

How can I address non-specific binding issues with MITF antibodies?

Non-specific binding with MITF antibodies can confound experimental results, but several methodological approaches can mitigate these issues. First, optimize antibody dilutions - start with the manufacturer's recommended range (e.g., 1:500-1:3000 for Western blotting with Proteintech's antibody), but perform a dilution series to identify the concentration that maximizes specific signal while minimizing background . Second, enhance blocking protocols by extending blocking time (1-2 hours at room temperature or overnight at 4°C) and testing different blocking agents (BSA, normal serum, or commercial blockers) appropriate for your application. Third, increase washing frequency and duration between antibody incubations, using buffers containing 0.1-0.5% Tween-20 or Triton X-100 to remove weakly bound antibodies. Fourth, for Western blotting specifically, use freshly prepared samples with protease inhibitors to prevent degradation products that may generate confusing bands, and consider gradient gels to better resolve MITF from similarly sized proteins. Fifth, pre-adsorb antibodies with tissues or cell lysates from species with which cross-reactivity is observed but not desired. Finally, if persistent non-specific binding occurs in ChIP experiments, increase the stringency of wash buffers or implement a pre-clearing step with protein A/G beads before adding the MITF antibody. Remember that MITF appears at 50-75 kDa range on Western blots depending on the isoform and post-translational modifications, so bands outside this range are likely non-specific .

How can MITF antibodies be used to study the relationship between MITF and the immune system in melanoma?

MITF antibodies provide crucial tools for investigating the complex interplay between MITF and the immune system in melanoma research through several advanced methodological approaches. First, multiplex immunofluorescence using MITF antibodies alongside immune cell markers can visualize spatial relationships between MITF-expressing melanoma cells and tumor-infiltrating lymphocytes within the tumor microenvironment . Second, chromatin immunoprecipitation sequencing (ChIP-seq) with MITF antibodies can identify direct transcriptional targets involved in immune regulation, including genes encoding coinhibitory receptors like PD-L1 and HVEM that MITF has been shown to modulate . Third, combining MITF immunoprecipitation with mass spectrometry can reveal protein-protein interactions between MITF and immune signaling pathway components. Fourth, cell sorting based on MITF expression levels followed by immune functional assays or transcriptome analysis can elucidate how varying MITF levels affect inflammatory secretome and immune cell recruitment or activation . Fifth, time-course studies using MITF antibodies can track changes in MITF expression during immune checkpoint inhibitor therapy, potentially identifying predictive biomarkers of response. Recent research has established MITF as "a central mediator in the regulation of immune responses in melanoma and other cancers," making these antibody-based approaches invaluable for understanding the molecular mechanisms through which MITF influences immune checkpoint inhibitory therapy resistance .

What methodologies are recommended for studying MITF's role in cellular differentiation and development?

To investigate MITF's role in cellular differentiation and development, researchers should employ multiple complementary antibody-based methodologies. First, implement time-course immunostaining studies during cellular differentiation processes (particularly in melanocytes and retinal pigment epithelium) using MITF antibodies alongside markers of differentiation status to correlate MITF expression patterns with developmental stages . Second, couple ChIP-seq analysis using validated MITF antibodies (applying the recommended protocols with 10 μl antibody and 10 μg chromatin) with RNA-seq to identify direct MITF target genes involved in differentiation pathways . Third, utilize co-immunoprecipitation with MITF antibodies followed by mass spectrometry to identify developmental stage-specific MITF protein interaction partners that may modulate its transcriptional activity. Fourth, perform simultaneous detection of multiple MITF isoforms using isoform-specific antibodies or antibodies recognizing common regions to track their differential expression during development - particularly important since at least 12 MITF isoforms exist with varying tissue expression patterns . Fifth, implement proximity ligation assays using MITF antibodies and antibodies against suspected interaction partners to visualize and quantify protein interactions in situ during developmental processes. Sixth, conduct ChIP-re-ChIP experiments using MITF antibodies in combination with antibodies against other transcription factors to identify genomic loci co-regulated by multiple factors during differentiation. These methodological approaches collectively provide a comprehensive framework for unraveling MITF's multifaceted roles in cellular differentiation and development.

How can MITF antibodies contribute to understanding the mechanisms of melanoma resistance to immunotherapy?

MITF antibodies offer powerful tools for deciphering melanoma immunotherapy resistance mechanisms through several sophisticated experimental approaches. First, perform immunohistochemistry or multiplex immunofluorescence with MITF antibodies on patient biopsies before treatment and upon resistance development to track changes in MITF expression patterns and heterogeneity as potential biomarkers of response . Second, conduct single-cell analyses combining MITF antibody-based detection with immune cell markers to characterize the relationship between MITF expression levels and immune cell infiltration or activation states at single-cell resolution. Third, implement in vitro models where MITF levels are experimentally manipulated, followed by co-culture with immune cells and subsequent antibody-based analysis of both melanoma and immune cell phenotypes to directly assess how MITF impacts immune cell function . Fourth, utilize ChIP-seq with MITF antibodies in immunotherapy-resistant versus sensitive tumors to identify differential MITF binding to genes involved in antigen presentation, inflammatory signaling, and immune checkpoint regulation . Fifth, combine MITF immunoprecipitation with proteomics to identify MITF-interacting proteins specifically in the context of immunotherapy resistance. Recent research has highlighted that "MITF functions downstream oncogenic pathways and microenvironment stimuli that restrain the immune responses" and may regulate "melanoma-specific antigen expression by interfering with the endolysosomal pathway, KARS1, and antigen processing and presentation" . These methodological approaches leveraging MITF antibodies can systematically dissect these proposed mechanisms of immunotherapy resistance.

What are the optimal storage conditions for maintaining MITF antibody integrity?

Proper storage of MITF antibodies is critical for maintaining their integrity and ensuring consistent experimental results over time. Store MITF antibodies at -20°C for long-term preservation, which is the standard recommendation across multiple manufacturers . For the Active Motif MITF antibody, the product is shipped in a buffer containing 70 mM Tris (pH 8), 105 mM NaCl, 31 mM glycine, 0.07 mM EDTA, 30% glycerol, and 0.035% sodium azide, which helps maintain stability during storage . To prevent degradation from repeated freeze-thaw cycles, aliquot MITF antibodies into single-use fractions upon receipt - this is particularly important for antibodies without stabilizing proteins like BSA in their formulation . Some MITF antibody preparations, such as the 20 μl sizes from certain manufacturers, may contain 0.1% BSA as an additional stabilizing agent . Keep all MITF antibody reagents on ice when not in storage to minimize degradation during laboratory handling . For day-to-day use, small working aliquots can be kept at 4°C for up to one week, but extended storage at this temperature is not recommended. Most MITF antibodies are guaranteed for 12 months from the date of receipt when stored properly, though actual shelf life may be longer when optimal storage conditions are maintained .

What dilution protocols should be followed for different MITF antibody applications?

Optimal dilution protocols for MITF antibodies vary significantly based on the specific application and the antibody source, requiring careful attention to manufacturer recommendations and experimental validation. For Western blotting applications, the Cell Signaling MITF (D5G7V) Rabbit mAb requires a 1:1000 dilution , while the Proteintech polyclonal antibody allows a broader range from 1:500 to 1:3000, giving researchers flexibility to optimize signal-to-noise ratio for their specific samples . For chromatin immunoprecipitation (ChIP) applications, more concentrated antibody preparations are typically needed, with recommended dilutions of 1:50 for both standard ChIP and ChIP-seq protocols using the Cell Signaling antibody . For immunoprecipitation applications using the Proteintech polyclonal antibody, use 0.5-4.0 μg of antibody for each 1.0-3.0 mg of total protein lysate . Flow cytometry applications for intracellular MITF detection require approximately 0.25 μg of antibody per 10^6 cells in a 100 μl suspension . When establishing a new application or working with a new biological system, perform a dilution series experiment to determine the optimal concentration that maximizes specific signal while minimizing background. For all applications, prepare antibody dilutions freshly in appropriate diluents containing 0.1-1% BSA or other stabilizing proteins and maintain at 4°C during the dilution process to preserve antibody activity.

How can I validate lot-to-lot consistency when working with MITF antibodies?

Validating lot-to-lot consistency of MITF antibodies is essential for ensuring experimental reproducibility in long-term research projects. Implement these methodological approaches to systematically assess new antibody lots: First, perform side-by-side Western blot analysis using both the previous and new antibody lots on identical positive control samples (such as A549 cells, Jurkat cells, or NIH/3T3 cells for certain antibodies) to compare band patterns, intensities, and molecular weights . Second, quantitatively analyze immunofluorescence signal intensity and localization patterns using standardized imaging parameters across antibody lots - MITF should consistently show nuclear localization in appropriate cell types . Third, maintain a frozen reference sample set that can be used to validate each new lot, ensuring consistent performance over time. Fourth, consider using recombinant MITF antibodies when available, as they offer "superior lot-to-lot consistency, continuous supply, and animal-free manufacturing" compared to conventional antibodies . Fifth, when performing ChIP or ChIP-seq experiments, validate each new lot using established positive and negative control primer sets that have been previously validated for MITF antibodies, such as those recommended by Active Motif . Finally, maintain detailed records of antibody performance across different lots, including optimal dilutions and incubation conditions, to track any subtle variations that might affect experimental outcomes. By systematically implementing these validation steps, researchers can minimize experimental variability arising from antibody inconsistency.

How can MITF antibodies be used to study Waardenburg syndrome and other MITF-associated disorders?

MITF antibodies provide essential tools for investigating Waardenburg syndrome type 2A, Tietz syndrome, and other MITF-associated disorders through several methodological approaches. First, conduct immunohistochemistry or immunofluorescence on patient-derived samples using validated MITF antibodies to assess expression patterns and subcellular localization of wild-type versus mutant MITF proteins . The nuclear localization typically observed with wild-type MITF may be altered with certain disease-causing mutations. Second, utilize Western blotting with MITF antibodies to quantify expression levels in patient samples compared to controls, following optimized protocols with appropriate dilutions (1:500-1:3000) . Third, implement co-immunoprecipitation studies using MITF antibodies to investigate how disease-causing mutations affect protein-protein interactions essential for MITF function. Fourth, perform ChIP-seq analysis in relevant cell types with both wild-type and mutant MITF to identify differential genomic binding patterns that may explain phenotypic differences between disorders . Fifth, develop in vitro models expressing disease-specific MITF mutations, then use antibody-based techniques to track how these mutations affect MITF phosphorylation by MAP kinase in response to c-kit activation, which normally upregulates MITF transcriptional activity . These approaches can help elucidate how specific mutations in the MITF gene lead to "auditory-pigmentary syndrome characterized by developmental defects in cells derived from neural crest," providing insights into pathogenic mechanisms and potentially identifying therapeutic targets .

What methodological approaches are recommended for using MITF antibodies in melanoma prognosis research?

For melanoma prognosis research using MITF antibodies, several methodological approaches can yield clinically relevant insights. First, perform immunohistochemistry on tissue microarrays containing melanoma samples with known clinical outcomes, using optimized MITF antibody protocols and quantitative scoring systems to correlate MITF expression patterns with patient survival and response to therapy . Second, implement multiplex immunofluorescence combining MITF antibodies with markers of tumor heterogeneity and immune infiltration to assess the relationship between MITF expression, tumor microenvironment, and clinical outcomes . Third, conduct longitudinal studies using MITF antibodies on sequential biopsies from the same patients to track changes in MITF expression during disease progression and treatment, potentially identifying dynamic biomarkers of therapeutic response. Fourth, utilize single-cell analysis techniques with MITF antibodies to characterize intratumoral heterogeneity of MITF expression, which has been identified as "one of the major hurdles for effective immunotherapy" . Fifth, perform chromatin immunoprecipitation with MITF antibodies followed by sequencing (ChIP-seq) on patient-derived samples to identify MITF binding patterns associated with different prognostic groups . These approaches can help elucidate MITF's role as "a central mediator in the regulation of immune responses in melanoma" and potentially establish MITF expression profiles as biomarkers for stratifying patients for specific therapeutic approaches, particularly immunotherapies .

How might MITF antibodies contribute to emerging research on extracellular vesicle-based diagnostics in melanoma?

MITF antibodies hold significant potential for advancing extracellular vesicle (EV)-based melanoma diagnostics through several innovative methodological approaches. First, implement immunocapture techniques using MITF antibodies to isolate and enrich EVs specifically derived from melanoma cells, which may contain MITF protein or MITF-regulated cargo molecules . Second, develop flow cytometry protocols using fluorescently labeled MITF antibodies to detect and quantify MITF-positive EVs in patient blood samples, potentially establishing a liquid biopsy approach for melanoma monitoring. Third, perform immunogold labeling with MITF antibodies for transmission electron microscopy to visualize and confirm the presence of MITF in isolated EVs at ultrastructural resolution. Fourth, create multiplexed antibody arrays incorporating MITF antibodies alongside antibodies against other melanoma markers to comprehensively profile the protein content of circulating EVs. Fifth, utilize MITF antibodies in proximity ligation assays to detect specific protein interactions within EVs that might serve as unique diagnostic signatures. These approaches could potentially leverage the understanding that MITF regulates "melanoma-specific antigen expression" and produces "an inflammatory secretome," components of which might be packaged into EVs . By methodically investigating MITF-related content in melanoma-derived EVs, researchers could develop non-invasive diagnostic tools that reflect both the MITF expression status of the tumor and its potential impact on immune responses, ultimately advancing personalized medicine approaches for melanoma management.

What considerations should guide researchers studying MITF's role in non-melanoma contexts?

When investigating MITF's roles beyond melanoma contexts, researchers should implement these methodological considerations: First, carefully select MITF antibodies validated for the specific tissue type under investigation, as MITF expression exhibits distinct patterns across different tissues . The polyclonal antibody 13092-1-AP, for example, has been validated in multiple tissue types including heart tissue . Second, comprehensively characterize the MITF isoform profile in your specific biological system, as at least 12 MITF isoforms have been identified with tissue-specific expression patterns . Third, optimize immunohistochemistry and immunofluorescence protocols specifically for your tissue of interest, as fixation requirements and antigen retrieval methods may differ significantly from those established for melanoma samples. Fourth, when conducting ChIP experiments in non-melanoma contexts, validate MITF binding to both known melanoma-associated targets and tissue-specific candidate genes to establish conservation and divergence of MITF function across tissues. Fifth, implement co-expression studies using MITF antibodies alongside tissue-specific transcription factor antibodies to understand the contextual regulatory networks. Sixth, consider MITF's emerging role in kidney cancer research, where "it also plays a key role," and in immune cells, where it functions as "a central mediator in the regulation of immune responses" . These methodological considerations will help elucidate the conserved versus context-specific functions of MITF across diverse biological systems, potentially revealing novel therapeutic targets for multiple diseases.

How can CRISPR-based approaches be combined with MITF antibodies to advance melanoma research?

The integration of CRISPR-based approaches with MITF antibody techniques offers powerful methodological strategies for advancing melanoma research. First, implement CRISPR-mediated tagging of endogenous MITF with fluorescent proteins or epitope tags, followed by validation with existing MITF antibodies to ensure the tagged protein maintains normal localization and function . Second, utilize CRISPR interference (CRISPRi) or CRISPR activation (CRISPRa) to modulate MITF expression levels, then apply MITF antibodies in techniques like ChIP-seq to identify how altered MITF levels affect genome-wide binding patterns and target gene expression . Third, perform CRISPR screens targeting MITF transcriptional networks, followed by MITF immunoprecipitation and mass spectrometry to identify context-specific protein interactions under different selective pressures. Fourth, develop CRISPR base editing approaches to introduce specific MITF mutations associated with Waardenburg syndrome or melanoma progression, then use MITF antibodies to characterize the functional consequences on protein stability, localization, and activity . Fifth, combine CRISPR-mediated knockout of immune regulatory genes with MITF antibody-based analyses to dissect the "multifaceted interplay between microphthalmia-associated transcription factor (MITF)...and the immune system" . These integrated approaches can systematically elucidate how MITF contributes to "melanoma cell plasticity and tumor heterogeneity, which are undoubtedly one of the major hurdles for effective immunotherapy," potentially leading to improved therapeutic strategies for melanoma and other MITF-associated diseases .

What spatial transcriptomics approaches can be combined with MITF antibody techniques for melanoma research?

Integrating spatial transcriptomics with MITF antibody techniques offers revolutionary methodological approaches for investigating the spatial context of MITF function in melanoma research. First, implement sequential immunofluorescence using MITF antibodies followed by spatial transcriptomics on the same tissue section to directly correlate MITF protein expression with transcriptional profiles at single-cell resolution across the tumor microenvironment . Second, perform multiplexed ion beam imaging (MIBI) or imaging mass cytometry (IMC) incorporating MITF antibodies alongside markers for immune cells, enabling quantitative spatial analysis of how MITF-expressing melanoma cells interact with specific immune cell populations . Third, utilize spatial transcriptomics data to identify region-specific MITF target genes, then validate these through chromatin immunoprecipitation with MITF antibodies on microdissected tissue regions . Fourth, combine laser capture microdissection of MITF-high versus MITF-low regions (identified through immunohistochemistry) with single-cell RNA sequencing to characterize transcriptional heterogeneity associated with varying MITF expression levels. Fifth, implement in situ hybridization for MITF mRNA followed by immunofluorescence with MITF antibodies on serial sections to investigate post-transcriptional regulation of MITF expression. These approaches can systematically address the research challenge that "MITF is also a key determinant of melanoma cell plasticity and tumor heterogeneity," providing unprecedented spatial context for understanding how MITF influences the tumor microenvironment and potentially affecting "the infiltration and/or activation of the immune cells" in melanoma .

What are the key differences between commercially available MITF antibodies in terms of validation and performance?

Commercial MITF antibodies show significant variability in validation extent and application performance, requiring careful evaluation for specific research needs. The Cell Signaling MITF (D5G7V) Rabbit mAb (catalog #12590) has been extensively validated for Western blotting (1:1000 dilution) and ChIP applications (1:50 dilution), with demonstrations of cross-reactivity across human, mouse, rat, hamster, and monkey samples . In contrast, the Active Motif MITF antibody (clone C5) focuses validation on ChIP-Seq, Western blotting, immunofluorescence, and EMSA applications, with specific validation for both human and mouse samples through positive and negative control primer sets for ChIP experiments . The Proteintech polyclonal antibody (13092-1-AP) offers broader application validation, including Western blotting, immunoprecipitation, flow cytometry, and ELISA, with published validation in at least 30 Western blotting studies and 5 immunofluorescence publications . Regarding epitope targets, the Active Motif antibody was raised against an N-terminal fragment of human MITF, while others target different regions, potentially affecting isoform recognition . Performance in detecting specific molecular weight ranges also varies, with the Cell Signaling antibody recognizing MITF at 50-75 kDa and the Proteintech antibody detecting it at 59-65 kDa . Buffer compositions differ significantly between products, with Active Motif providing detailed buffer information (70 mM Tris pH 8, 105 mM NaCl, 31 mM glycine, 0.07 mM EDTA, 30% glycerol, 0.035% sodium azide) . When selecting an MITF antibody, researchers should prioritize products with validation data specific to their application of interest and consider reaching out to manufacturers for unpublished validation data in similar experimental systems.

What are the recommended best practices for designing experiments with MITF antibodies?

Designing robust experiments with MITF antibodies requires attention to several methodological best practices. First, select antibodies based on specific experimental requirements - consider whether you need isoform specificity, which applications you'll perform, and which species you're working with . Second, always include proper controls: positive controls (melanoma cell lines or tissues with known MITF expression), negative controls (tissues or cells without MITF expression), and technical controls (isotype antibodies, peptide competition) . Third, optimize protocols specifically for MITF detection - dilution ratios, incubation times, and buffer compositions should be systematically tested and standardized for your specific experimental system . Fourth, validate MITF antibody performance in your hands through preliminary experiments before proceeding to complex or resource-intensive studies. Fifth, consider the biological context - MITF expression and function vary significantly across cell types and can be modulated by microenvironmental factors, particularly in melanoma where MITF functions downstream of oncogenic pathways . Sixth, implement complementary approaches when possible, such as combining protein detection with mRNA analysis or using multiple antibodies targeting different MITF epitopes to confirm findings. Seventh, thoroughly document all experimental conditions, including antibody catalog numbers, lot numbers, dilutions, and incubation parameters to ensure reproducibility. Finally, when studying MITF in relation to immune responses, design experiments that capture the "multifaceted interplay between microphthalmia-associated transcription factor (MITF)...and the immune system," which requires careful consideration of both tumor cell and immune cell characteristics .

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.