Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 155(5): 1022-33, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24267888

ABSTRACT

Sequence polymorphisms linked to human diseases and phenotypes in genome-wide association studies often affect noncoding regions. A SNP within an intron of the gene encoding Interferon Regulatory Factor 4 (IRF4), a transcription factor with no known role in melanocyte biology, is strongly associated with sensitivity of skin to sun exposure, freckles, blue eyes, and brown hair color. Here, we demonstrate that this SNP lies within an enhancer of IRF4 transcription in melanocytes. The allele associated with this pigmentation phenotype impairs binding of the TFAP2A transcription factor that, together with the melanocyte master regulator MITF, regulates activity of the enhancer. Assays in zebrafish and mice reveal that IRF4 cooperates with MITF to activate expression of Tyrosinase (TYR), an essential enzyme in melanin synthesis. Our findings provide a clear example of a noncoding polymorphism that affects a phenotype by modulating a developmental gene regulatory network.


Subject(s)
Interferon Regulatory Factors/metabolism , Polymorphism, Single Nucleotide , Animals , Base Sequence , Enhancer Elements, Genetic , Humans , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/genetics , Melanocytes/metabolism , Mice , Molecular Sequence Data , Pigmentation , Signal Transduction , Transcription Factor AP-2/chemistry , Transcription Factor AP-2/metabolism , Zebrafish
2.
Mol Cell ; 79(3): 472-487.e10, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32531202

ABSTRACT

It is widely assumed that decreasing transcription factor DNA-binding affinity reduces transcription initiation by diminishing occupancy of sequence-specific regulatory elements. However, in vivo transcription factors find their binding sites while confronted with a large excess of low-affinity degenerate motifs. Here, using the melanoma lineage survival oncogene MITF as a model, we show that low-affinity binding sites act as a competitive reservoir in vivo from which transcription factors are released by mitogen-activated protein kinase (MAPK)-stimulated acetylation to promote increased occupancy of their regulatory elements. Consequently, a low-DNA-binding-affinity acetylation-mimetic MITF mutation supports melanocyte development and drives tumorigenesis, whereas a high-affinity non-acetylatable mutant does not. The results reveal a paradoxical acetylation-mediated molecular clutch that tunes transcription factor availability via genome-wide redistribution and couples BRAF to tumorigenesis. Our results further suggest that p300/CREB-binding protein-mediated transcription factor acetylation may represent a common mechanism to control transcription factor availability.


Subject(s)
Gene Expression Regulation, Neoplastic , Genome , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , Protein Processing, Post-Translational , Skin Neoplasms/genetics , Acetylation , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Tumor , Conserved Sequence , Enhancer Elements, Genetic , Female , Heterografts , Humans , Male , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Microphthalmia-Associated Transcription Factor/chemistry , Microphthalmia-Associated Transcription Factor/metabolism , Nucleotide Motifs , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Sequence Alignment , Sequence Homology, Amino Acid , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Zebrafish
3.
Traffic ; 25(4): e12933, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38600522

ABSTRACT

Macroautophagy/autophagy is an essential catabolic process that targets a wide variety of cellular components including proteins, organelles, and pathogens. ATG7, a protein involved in the autophagy process, plays a crucial role in maintaining cellular homeostasis and can contribute to the development of diseases such as cancer. ATG7 initiates autophagy by facilitating the lipidation of the ATG8 proteins in the growing autophagosome membrane. The noncanonical isoform ATG7(2) is unable to perform ATG8 lipidation; however, its cellular regulation and function are unknown. Here, we uncovered a distinct regulation and function of ATG7(2) in contrast with ATG7(1), the canonical isoform. First, affinity-purification mass spectrometry analysis revealed that ATG7(2) establishes direct protein-protein interactions (PPIs) with metabolic proteins, whereas ATG7(1) primarily interacts with autophagy machinery proteins. Furthermore, we identified that ATG7(2) mediates a decrease in metabolic activity, highlighting a novel splice-dependent function of this important autophagy protein. Then, we found a divergent expression pattern of ATG7(1) and ATG7(2) across human tissues. Conclusively, our work uncovers the divergent patterns of expression, protein interactions, and function of ATG7(2) in contrast to ATG7(1). These findings suggest a molecular switch between main catabolic processes through isoform-dependent expression of a key autophagy gene.


Subject(s)
Autophagy , Energy Metabolism , Humans , Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Protein Isoforms/metabolism
4.
PLoS Genet ; 18(5): e1010207, 2022 05.
Article in English | MEDLINE | ID: mdl-35580127

ABSTRACT

In developing melanocytes and in melanoma cells, multiple paralogs of the Activating-enhancer-binding Protein 2 family of transcription factors (TFAP2) contribute to expression of genes encoding pigmentation regulators, but their interaction with Microphthalmia transcription factor (MITF), a master regulator of these cells, is unclear. Supporting the model that TFAP2 facilitates MITF's ability to activate expression of pigmentation genes, single-cell seq analysis of zebrafish embryos revealed that pigmentation genes are only expressed in the subset of mitfa-expressing cells that also express tfap2 paralogs. To test this model in SK-MEL-28 melanoma cells we deleted the two TFAP2 paralogs with highest expression, TFAP2A and TFAP2C, creating TFAP2 knockout (TFAP2-KO) cells. We then assessed gene expression, chromatin accessibility, binding of TFAP2A and of MITF, and the chromatin marks H3K27Ac and H3K27Me3 which are characteristic of active enhancers and silenced chromatin, respectively. Integrated analyses of these datasets indicate TFAP2 paralogs directly activate enhancers near genes enriched for roles in pigmentation and proliferation, and directly repress enhancers near genes enriched for roles in cell adhesion. Consistently, compared to WT cells, TFAP2-KO cells proliferate less and adhere to one another more. TFAP2 paralogs and MITF co-operatively activate a subset of enhancers, with the former necessary for MITF binding and chromatin accessibility. By contrast, TFAP2 paralogs and MITF do not appear to co-operatively inhibit enhancers. These studies reveal a mechanism by which TFAP2 profoundly influences the set of genes activated by MITF, and thereby the phenotype of pigment cells and melanoma cells.


Subject(s)
Melanoma , Microphthalmos , Animals , Cell Proliferation/genetics , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Developmental , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Microphthalmos/genetics , Pigmentation/genetics , Zebrafish/genetics , Zebrafish/metabolism
5.
Am J Hum Genet ; 108(9): 1631-1646, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34293285

ABSTRACT

Although expression quantitative trait loci (eQTLs) have been powerful in identifying susceptibility genes from genome-wide association study (GWAS) findings, most trait-associated loci are not explained by eQTLs alone. Alternative QTLs, including DNA methylation QTLs (meQTLs), are emerging, but cell-type-specific meQTLs using cells of disease origin have been lacking. Here, we established an meQTL dataset by using primary melanocytes from 106 individuals and identified 1,497,502 significant cis-meQTLs. Multi-QTL colocalization with meQTLs, eQTLs, and mRNA splice-junction QTLs from the same individuals together with imputed methylome-wide and transcriptome-wide association studies identified candidate susceptibility genes at 63% of melanoma GWAS loci. Among the three molecular QTLs, meQTLs were the single largest contributor. To compare melanocyte meQTLs with those from malignant melanomas, we performed meQTL analysis on skin cutaneous melanomas from The Cancer Genome Atlas (n = 444). A substantial proportion of meQTL probes (45.9%) in primary melanocytes is preserved in melanomas, while a smaller fraction of eQTL genes is preserved (12.7%). Integration of melanocyte multi-QTLs and melanoma meQTLs identified candidate susceptibility genes at 72% of melanoma GWAS loci. Beyond GWAS annotation, meQTL-eQTL colocalization in melanocytes suggested that 841 unique genes potentially share a causal variant with a nearby methylation probe in melanocytes. Finally, melanocyte trans-meQTLs identified a hotspot for rs12203592, a cis-eQTL of a transcription factor, IRF4, with 131 candidate target CpGs. Motif enrichment and IRF4 ChIP-seq analysis demonstrated that these target CpGs are enriched in IRF4 binding sites, suggesting an IRF4-mediated regulatory network. Our study highlights the utility of cell-type-specific meQTLs.


Subject(s)
Gene Regulatory Networks , Interferon Regulatory Factors/genetics , Melanocytes/metabolism , Melanoma/genetics , Quantitative Trait Loci , Skin Neoplasms/genetics , Alleles , Atlases as Topic , Chromatin/chemistry , Chromatin/metabolism , Chromosome Mapping , DNA Methylation , Gene Expression Regulation , Genetic Predisposition to Disease , Genome, Human , Genome-Wide Association Study , Humans , Infant, Newborn , Interferon Regulatory Factors/metabolism , Male , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Primary Cell Culture , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcriptome
6.
Nucleic Acids Res ; 48(2): 934-948, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31777941

ABSTRACT

Interrupted dimeric coiled coil segments are found in a broad range of proteins and generally confer selective functional properties such as binding to specific ligands. However, there is only one documented case of a basic-helix-loop-helix leucine zipper transcription factor-microphthalmia-associated transcription factor (MITF)-in which an insertion of a three-residue stammer serves as a determinant of conditional partner selectivity. To unravel the molecular principles of this selectivity, we have analyzed the high-resolution structures of stammer-containing MITF and an engineered stammer-less MITF variant, which comprises an uninterrupted symmetric coiled coil. Despite this fundamental difference, both MITF structures reveal identical flanking in-phase coiled coil arrangements, gained by helical over-winding and local asymmetry in wild-type MITF across the stammer region. These conserved structural properties allow the maintenance of a proper functional readout in terms of nuclear localization and binding to specific DNA-response motifs regardless of the presence of the stammer. By contrast, MITF heterodimer formation with other bHLH-Zip transcription factors is only permissive when both factors contain either the same type of inserted stammer or no insert. Our data illustrate a unique principle of conditional partner selectivity within the wide arsenal of transcription factors with specific partner-dependent functional readouts.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Cell Nucleus/chemistry , Microphthalmia-Associated Transcription Factor/chemistry , Protein Conformation , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Ligands , Mice , Microphthalmia-Associated Transcription Factor/genetics , Protein Binding , Protein Domains/genetics , Protein Multimerization
7.
Exp Dermatol ; 30(8): 1033-1050, 2021 08.
Article in English | MEDLINE | ID: mdl-34003523

ABSTRACT

Melanocytes originate in the neural crest as precursor cells which then migrate and proliferate to reach their destination where they differentiate into pigment-producing cells. Melanocytes not only determine the colour of hair, skin and eyes but also protect against the harmful effects of UV irradiation. The establishment of the melanocyte lineage is regulated by a defined set of transcription factors and signalling pathways that direct the specific gene expression programmes underpinning melanoblast specification, survival, migration, proliferation and differentiation. In addition, epigenetic modifiers and replacement histones play key roles in regulating gene expression and its timing during the different steps of this process. Here, we discuss the evidence for the role of epigenetic regulators in melanocyte development and function and how they interact with transcription factors and signalling pathways to establish and maintain this important cell lineage.


Subject(s)
Epigenesis, Genetic , Homeostasis/genetics , Melanocytes/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Cell Lineage , Humans , Mice
8.
Proc Natl Acad Sci U S A ; 115(37): E8668-E8677, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150413

ABSTRACT

The close integration of the MAPK, PI3K, and WNT signaling pathways underpins much of development and is deregulated in cancer. In principle, combinatorial posttranslational modification of key lineage-specific transcription factors would be an effective means to integrate critical signaling events. Understanding how this might be achieved is central to deciphering the impact of microenvironmental cues in development and disease. The microphthalmia-associated transcription factor MITF plays a crucial role in the development of melanocytes, the retinal pigment epithelium, osteoclasts, and mast cells and acts as a lineage survival oncogene in melanoma. MITF coordinates survival, differentiation, cell-cycle progression, cell migration, metabolism, and lysosome biogenesis. However, how the activity of this key transcription factor is controlled remains poorly understood. Here, we show that GSK3, downstream from both the PI3K and Wnt pathways, and BRAF/MAPK signaling converges to control MITF nuclear export. Phosphorylation of the melanocyte MITF-M isoform in response to BRAF/MAPK signaling primes for phosphorylation by GSK3, a kinase inhibited by both PI3K and Wnt signaling. Dual phosphorylation, but not monophosphorylation, then promotes MITF nuclear export by activating a previously unrecognized hydrophobic export signal. Nonmelanocyte MITF isoforms exhibit poor regulation by MAPK signaling, but instead their export is controlled by mTOR. We uncover here an unanticipated mode of MITF regulation that integrates the output of key developmental and cancer-associated signaling pathways to gate MITF flux through the import-export cycle. The results have significant implications for our understanding of melanoma progression and stem cell renewal.


Subject(s)
Cell Nucleus/metabolism , Glycogen Synthase Kinase 3/metabolism , MAP Kinase Signaling System , Microphthalmia-Associated Transcription Factor/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cells, Cultured , HeLa Cells , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Mutation , Phosphorylation , Protein Binding
9.
Clin Genet ; 98(4): 353-364, 2020 10.
Article in English | MEDLINE | ID: mdl-33111345

ABSTRACT

Mutations in more than 150 genes are responsible for inherited hearing loss, with thousands of different, severe causal alleles that vary among populations. The Israeli Jewish population includes communities of diverse geographic origins, revealing a wide range of deafness-associated variants and enabling clinical characterization of the associated phenotypes. Our goal was to identify the genetic causes of inherited hearing loss in this population, and to determine relationships among genotype, phenotype, and ethnicity. Genomic DNA samples from informative relatives of 88 multiplex families, all of self-identified Jewish ancestry, with either non-syndromic or syndromic hearing loss, were sequenced for known and candidate deafness genes using the HEar-Seq gene panel. The genetic causes of hearing loss were identified for 60% of the families. One gene was encountered for the first time in human hearing loss: ATOH1 (Atonal), a basic helix-loop-helix transcription factor responsible for autosomal dominant progressive hearing loss in a five-generation family. Our results show that genomic sequencing with a gene panel dedicated to hearing loss is effective for genetic diagnoses in a diverse population. Comprehensive sequencing enables well-informed genetic counseling and clinical management by medical geneticists, otolaryngologists, audiologists, and speech therapists and can be integrated into newborn screening for deafness.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Deafness/genetics , Genetic Predisposition to Disease , Hearing Loss/genetics , Adolescent , Adult , Child , Child, Preschool , Deafness/epidemiology , Deafness/pathology , Female , Genetic Association Studies , Hearing Loss/epidemiology , Hearing Loss/pathology , Humans , Israel/epidemiology , Jews/genetics , Male , Pedigree , Young Adult
10.
Acta Derm Venereol ; 100(11): adv00139, 2020 Jun 03.
Article in English | MEDLINE | ID: mdl-32346747

ABSTRACT

Cutaneous melanoma arises from melanocytes following genetic, epigenetic and allogenetic (i.e. other than epi/genetic) modifications. An estimated 10% of cutaneous melanoma cases are due to inherited variants or de novo mutations in approximately 20 genes, found using linkage, next-generation sequencing and association studies. Based on these studies, 3 classes of predisposing melanoma genes have been defined based on the frequency of the variants in the general population and lifetime risk of developing a melanoma: (i) ultra-rare variants with a high risk, (ii) rare with a moderate risk, and (iii) frequent variants with a low risk. Most of the proteins encoded by these genes have been shown to be involved in melanoma initiation, including proliferation and senescence bypass. This paper reviews the role(s) of these genes in the transformation of melanocytes into melanoma. It also describes their function in the establishment and renewal of melanocytes and the biology of pigment cells, if known.


Subject(s)
Biomarkers, Tumor/genetics , Melanocytes/pathology , Melanoma/genetics , Mutation , Skin Neoplasms/genetics , Animals , Cell Lineage , Genetic Predisposition to Disease , Humans , Melanins/metabolism , Melanocytes/metabolism , Melanoma/ethnology , Melanoma/metabolism , Melanoma/pathology , Melanosomes/metabolism , Melanosomes/pathology , Mutation Rate , Phenotype , Risk Assessment , Risk Factors , Skin Neoplasms/ethnology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , White People/genetics
11.
Genes Dev ; 26(23): 2647-58, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23207919

ABSTRACT

Microphthalmia-associated transcription factor (MITF) is a master regulator of melanocyte development and an important oncogene in melanoma. MITF heterodimeric assembly with related basic helix-loop-helix leucine zipper transcription factors is highly restricted, and its binding profile to cognate DNA sequences is distinct. Here, we determined the crystal structure of MITF in its apo conformation and in the presence of two related DNA response elements, the E-box and M-box. In addition, we investigated mouse and human Mitf mutations to dissect the functional significance of structural features. Owing to an unusual three-residue shift in the leucine zipper register, the MITF homodimer shows a marked kink in one of the two zipper helices to allow an out-of-register assembly. Removal of this insertion relieves restricted heterodimerization by MITF and permits assembly with the transcription factor MAX. Binding of MITF to the M-box motif is mediated by an unusual nonpolar interaction by Ile212, a residue that is mutated in mice and humans with Waardenburg syndrome. As several related transcription factors have low affinity for the M-box sequence, our analysis unravels how these proteins discriminate between similar target sequences. Our data provide a rational basis for targeting MITF in the treatment of important hereditary diseases and cancer.


Subject(s)
Leucine Zippers , Microphthalmia-Associated Transcription Factor/chemistry , Microphthalmia-Associated Transcription Factor/metabolism , Models, Molecular , Amino Acid Sequence , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Enhancer Elements, Genetic/genetics , Humans , Leucine Zippers/genetics , Mice , Microphthalmia-Associated Transcription Factor/genetics , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Waardenburg Syndrome/genetics
12.
Pharmacoepidemiol Drug Saf ; 28(4): 471-478, 2019 04.
Article in English | MEDLINE | ID: mdl-30565786

ABSTRACT

PURPOSE: Increased expression of Vacuolar-type H+ ATPases (V-ATPases), in the plasma membrane of cancer cells has been suggested to contribute to the development of aggressive cancer phenotypes by promoting acidic tumor microenvironments. Accumulating data suggest that proton pump inhibitors (PPIs) may elicit a chemopreventive effect via V-ATPase inhibition in some cancers, but evidence is still limited. Therefore, we aimed to explore a potential preventive role of PPIs in this study. METHODS: In this population-based case-control study, we identified incident cases of breast cancer (n = 1739), prostate cancer (n = 1897), and malignant melanoma (n = 385) in Iceland between 2005 and 2014 from the Icelandic Cancer Registry. We assessed varying levels of PPI use through record linkages to the Icelandic Medicines Registry. For each case, we selected up to 10 age-matched, sex-matched, and calendar-matched population controls using risk-set sampling. Using conditional logistic regression, we calculated odds ratios (ORs) and 95% confidence intervals (CIs) controlling for NSAID use. RESULTS: Adjusted ORs associated with ever use of PPIs were 1.03 (95% CI: 0.92-1.16) for breast cancer, 1.12 (95% CI: 1.00-1.25) for prostate cancer, and 0.84 (95% CI: 0.69-1.12) for malignant melanoma. Analyses of high use of PPIs (≥1000 DDDs) yielded ORs of 0.97 (95% CI: 0.78-1.19), 1.20 (0.99-1.47), and 0.59 (0.40-1.13) for breast cancer, prostate cancer, and malignant melanoma, respectively. Analyses of cumulative exposure to PPIs did not support a dose-response relationship for any of the three cancer types. CONCLUSIONS: Our findings do not support a chemopreventive effect of PPI use on breast cancer, prostate cancer, or malignant melanoma.


Subject(s)
Breast Neoplasms/epidemiology , Melanoma/epidemiology , Prostatic Neoplasms/epidemiology , Proton Pump Inhibitors/administration & dosage , Skin Neoplasms/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/prevention & control , Case-Control Studies , Female , Humans , Iceland/epidemiology , Logistic Models , Male , Melanoma/prevention & control , Middle Aged , Odds Ratio , Prostatic Neoplasms/prevention & control , Registries/statistics & numerical data , Risk Factors , Skin Neoplasms/prevention & control , Tumor Microenvironment/drug effects , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism , Young Adult
13.
J Cell Sci ; 128(15): 2938-50, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26092939

ABSTRACT

The v-ATPase is a fundamental eukaryotic enzyme that is central to cellular homeostasis. Although its impact on key metabolic regulators such as TORC1 is well documented, our knowledge of mechanisms that regulate v-ATPase activity is limited. Here, we report that the Drosophila transcription factor Mitf is a master regulator of this holoenzyme. Mitf directly controls transcription of all 15 v-ATPase components through M-box cis-sites and this coordinated regulation affects holoenzyme activity in vivo. In addition, through the v-ATPase, Mitf promotes the activity of TORC1, which in turn negatively regulates Mitf. We provide evidence that Mitf, v-ATPase and TORC1 form a negative regulatory loop that maintains each of these important metabolic regulators in relative balance. Interestingly, direct regulation of v-ATPase genes by human MITF also occurs in cells of the melanocytic lineage, showing mechanistic conservation in the regulation of the v-ATPase by MITF family proteins in fly and mammals. Collectively, this evidence points to an ancient module comprising Mitf, v-ATPase and TORC1 that serves as a dynamic modulator of metabolism for cellular homeostasis.


Subject(s)
Drosophila Proteins/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Transcription Factors/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Drosophila , Enzyme Activation , Homeostasis/physiology , Humans , Melanocytes/metabolism , Melanoma/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering , Transcription, Genetic/genetics , Vacuolar Proton-Translocating ATPases/metabolism
14.
Hum Mol Genet ; 22(21): 4357-67, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23787126

ABSTRACT

The basic-helix-loop-helix-leucine zipper (bHLHZip) protein MITF (microphthalmia-associated transcription factor) is a master regulator of melanocyte development. Mutations in the MITF have been found in patients with the dominantly inherited hypopigmentation and deafness syndromes Waardenburg syndrome type 2A (WS2A) and Tietz syndrome (TS). Additionally, both somatic and germline mutations have been found in MITF in melanoma patients. Here, we characterize the DNA-binding and transcription activation properties of 24 MITF mutations found in WS2A, TS and melanoma patients. We show that most of the WS2A and TS mutations fail to bind DNA and activate expression from melanocyte-specific promoters. Some of the mutations, especially R203K and S298P, exhibit normal activity and may represent neutral variants. Mutations found in melanomas showed normal DNA-binding and minor variations in transcription activation properties; some showed increased potential to form colonies. Our results provide molecular insights into how mutations in a single gene can lead to such different phenotypes.


Subject(s)
Albinism, Oculocutaneous/genetics , Deafness/genetics , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Waardenburg Syndrome/genetics , Adolescent , Adult , Albinism, Oculocutaneous/metabolism , Albinism, Oculocutaneous/pathology , Binding Sites , Child , Child, Preschool , Deafness/metabolism , Deafness/pathology , Female , Genetic Variation , HEK293 Cells , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Mutation, Missense , Promoter Regions, Genetic , Transcriptional Activation , Transfection , Waardenburg Syndrome/metabolism , Waardenburg Syndrome/pathology , Young Adult
15.
Front Immunol ; 15: 1339325, 2024.
Article in English | MEDLINE | ID: mdl-38444862

ABSTRACT

Introduction: The microphthalmia transcription factor Mitf has been shown to regulate B cell activation and tolerance. However, the underlying B cell-specific mechanisms responsible, and those that distinguish Mitf from closely related Mitf/TFE (MiT) transcription factors Tfe3, Tfeb, and Tfec, remain obscure. Methods: Two complementary mouse models of Mitf and MiT deficiency were used: the Mitfmi-vga9/mi-vga9 systemic loss-of-function mutation, and B-cell specific MiT family inactivation via transgenic expression of a trans-dominant negative (TDN) protein (TDN-B). These models were employed to identify MiT family candidate target genes and pathways. Results: Both models displayed spontaneous splenomegaly coincident with elevated plasma cell numbers, autoantibody titers, and proteinuria. These abnormalities appeared dependent on T helper cells, but independent of other non-B cell intrinsic effects of systemic Mitf inactivation. MiT inactivation in B cells augmented aspects of lupus-like autoimmune disease on the C57BL/6-Faslpr/lpr background. In both models, RNAseq of ex vivo resting B cells showed transcriptional upregulation of genes that control cell cycle, germinal center responses, and plasma cell differentiation. Among the genes strongly upregulated in both models were Socs6, Isp53 (Baiap1), S1pR2, and IgG2b/c. Mitf null B cells, but not TDN-B cells, showed evidence of type I interferon dysregulation. Discussion: These studies clarify Mitf's role as 1) a key regulator of a B cell intrinsic germinal center program that influences self-tolerance through novel target genes, and 2) a regulator of systemic inflammatory processes that can impact the B cell microenvironment. This distinction of Mitf's function from that of related MiT transcription factors advances our understanding of B cell regulation and autoimmunity.


Subject(s)
B-Lymphocytes , Germinal Center , Animals , Mice , Gene Expression , Homeostasis , Mice, Inbred C57BL
16.
FEBS J ; 290(12): 3059-3075, 2023 06.
Article in English | MEDLINE | ID: mdl-35305060

ABSTRACT

Hedgehog signalling is essential for development, crucial for normal anatomical arrangement and activated during tissue damage repair. Dysregulation of hedgehog signalling is associated with cancer, developmental disorders and other diseases including osteoarthritis (OA). The hedgehog gene was first discovered in Drosophila melanogaster, and the pathway is evolutionarily conserved in most animals. Although there are several hedgehog ligands with different protein expression patterns, they share a common plasma membrane receptor, Patched1 and hedgehog signalling pathway activation is transduced through the G-protein-coupled receptor-like protein Smoothened (SMO) and downstream effectors. Functional assays revealed that activation of SMO is dependent on sterol binding, and cholesterol was observed bound to SMO in crystallography experiments. In vertebrates, hedgehog signalling coordinates endochondral ossification and balances osteoblast and osteoclast activation to maintain homeostasis. A recently discovered mutation of SMO in humans (SMOR173C ) is predicted to alter cholesterol binding and is associated with a higher risk of hip OA. Functional studies in mice and human tissue analysis provide evidence that hedgehog signalling is pathologically activated in chondrocytes of osteoarthritic cartilage.


Subject(s)
Drosophila Proteins , Osteoarthritis , Animals , Humans , Mice , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Cholesterol , Osteoarthritis/genetics
17.
bioRxiv ; 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37786677

ABSTRACT

MITF, a basic-Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. To explore MITF regulation and its role in melanoma, we conducted a genetic screen for suppressors of the Mitf-associated pigmentation phenotype. An intragenic Mitf mutation was identified, leading to termination of MITF at the K316 SUMOylation site and loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. Interestingly, as a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability and ensuring an active nuclear MITF supply. Interactions between K316 SUMOylation and S409 phosphorylation sites across monomers largely explain the observed effects. Notably, the recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409, unraveling a novel regulatory mechanism with unexpected disease insights.

18.
Nat Commun ; 14(1): 6051, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37770430

ABSTRACT

The ability of transcription factors to discriminate between different classes of binding sites associated with specific biological functions underpins effective gene regulation in development and homeostasis. How this is achieved is poorly understood. The microphthalmia-associated transcription factor MITF is a lineage-survival oncogene that plays a crucial role in melanocyte development and melanoma. MITF suppresses invasion, reprograms metabolism and promotes both proliferation and differentiation. How MITF distinguishes between differentiation and proliferation-associated targets is unknown. Here we show that compared to many transcription factors MITF exhibits a very long residence time which is reduced by p300/CBP-mediated MITF acetylation at K206. While K206 acetylation also decreases genome-wide MITF DNA-binding affinity, it preferentially directs DNA binding away from differentiation-associated CATGTG motifs toward CACGTG elements. The results reveal an acetylation-mediated switch that suppresses differentiation and provides a mechanistic explanation of why a human K206Q MITF mutation is associated with Waardenburg syndrome.


Subject(s)
Melanoma , Microphthalmia-Associated Transcription Factor , Humans , Cell Line, Tumor , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Acetylation , Melanoma/genetics , Melanoma/metabolism , Melanocytes/metabolism
19.
Sci Rep ; 12(1): 10333, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35725745

ABSTRACT

Autophagy is a housekeeping mechanism tasked with eliminating misfolded proteins and damaged organelles to maintain cellular homeostasis. Autophagy deficiency results in increased oxidative stress, DNA damage and chronic cellular injury. Among the core genes in the autophagy machinery, ATG7 is required for autophagy initiation and autophagosome formation. Based on the analysis of an extended pedigree of familial cholangiocarcinoma, we determined that all affected family members had a novel germline mutation (c.2000C>T p.Arg659* (p.R659*)) in ATG7. Somatic deletions of ATG7 were identified in the tumors of affected individuals. We applied linked-read sequencing to one tumor sample and demonstrated that the ATG7 somatic deletion and germline mutation were located on distinct alleles, resulting in two hits to ATG7. From a parallel population genetic study, we identified a germline polymorphism of ATG7 (c.1591C>G p.Asp522Glu (p.D522E)) associated with increased risk of cholangiocarcinoma. To characterize the impact of these germline ATG7 variants on autophagy activity, we developed an ATG7-null cell line derived from the human bile duct. The mutant p.R659* ATG7 protein lacked the ability to lipidate its LC3 substrate, leading to complete loss of autophagy and increased p62 levels. Our findings indicate that germline ATG7 variants have the potential to impact autophagy function with implications for cholangiocarcinoma development.


Subject(s)
Autophagy-Related Protein 7 , Bile Duct Neoplasms , Cholangiocarcinoma , RNA-Binding Proteins , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic , Cholangiocarcinoma/genetics , Germ Cells/metabolism , Humans , RNA-Binding Proteins/genetics
20.
Pigment Cell Melanoma Res ; 34(1): 13-27, 2021 01.
Article in English | MEDLINE | ID: mdl-32846025

ABSTRACT

The microphthalmia-associated transcription factor (MITF) is at the core of melanocyte and melanoma fate specification. The related factors TFEB and TFE3 have been shown to be instrumental for transcriptional regulation of genes involved in lysosome biogenesis and autophagy, cellular processes important for mediating nutrition signals and recycling of cellular materials, in many cell types. The MITF, TFEB, TFE3, and TFEC proteins are highly related. They share many structural and functional features and are targeted by the same signaling pathways. However, the existence of several isoforms of each factor and the increasing number of residues shown to be post-translationally modified by various signaling pathways poses a difficulty in indexing amino acid residues in different isoforms across the different proteins. Here, we provide a resource manual to cross-reference amino acids and post-translational modifications in all isoforms of the MiT-TFE family in humans, mice, and zebrafish and summarize the protein accession numbers for each isoform of these factors in the different genomic databases. This will facilitate future studies on the signaling pathways that regulate different isoforms of the MiT-TFE transcription factor family.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Microphthalmia-Associated Transcription Factor/chemistry , Microphthalmia-Associated Transcription Factor/metabolism , Protein Processing, Post-Translational , Animals , Humans , Mice , Protein Isoforms , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL