Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Anticancer Drugs ; 35(1): 46-54, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37449977

RESUMEN

Protein degraders are currently under rapid development as a promising modality for drug discovery. They are compounds that orchestrate interactions between a target protein and an E3 ubiquitin ligase, prompting intracellular protein degradation through proteasomal pathway. More protein degraders identification will greatly promote the development of this field. BAG3 is widely recognized as an excellent therapeutic target in cancer treatments. Exploring protein degraders that target BAG3 degradation has profound implications. Herein, molecular docking was applied to assess binding energy between 81 clinical phase I kinase inhibitors and BAG3. BAG3 protein and mRNA level were detected by western blot and quantitative real-time PCR. CCK8 assay and colony formation assay were applied to detect the cell viability and proliferation rate. Cell death was accessed using flow cytometry combined with PI and Annexin V double staining. AZD7762, a Chk1 kinase inhibitor, was identified to induce BAG3 degradation in a ubiquitin-proteasome pathway. AZD7762-induced BAG3 degradation was not dependent on Chk1 expression or activity. CRBN, an E3 ligase, was identified to bind to BAG3 and mediated BAG3 ubiquitination in the presence of AZD7762. By targeting Chk1 and BAG3, two ideal therapeutic targets in cancer treatment, AZD7762 would be a powerful chemotherapy agent in the future.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Ubiquitina-Proteína Ligasas , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Línea Celular Tumoral , Simulación del Acoplamiento Molecular , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(43): 21732-21738, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31594848

RESUMEN

Endoplasmic reticulum (ER) stress plays an important role in metabolic diseases like obesity and type 2 diabetes mellitus (T2DM), although the underlying mechanisms and regulatory pathways remain to be elucidated. Here, we induced chronic low-grade ER stress in lean mice to levels similar to those in high-fat diet (HFD)-fed obese mice and found that it promoted hyperglycemia due to enhanced hepatic gluconeogenesis. Mechanistically, sustained ER stress up-regulated the deubiquitinating enzyme ubiquitin-specific peptidase 14 (USP14), which increased the stability and levels of 3',5'-cyclic monophosphate-responsive element binding (CREB) protein (CBP) to enhance glucagon action and hepatic gluconeogenesis. Exogenous overexpression of USP14 in the liver significantly increased hepatic glucose output. Consistent with this, liver-specific knockdown of USP14 abrogated the effects of ER stress on glucose metabolism, and also improved hyperglycemia and glucose intolerance in obese mice. In conclusion, our findings show a mechanism underlying ER stress-induced disruption of glucose homeostasis, and present USP14 as a potential therapeutic target against T2DM.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Glucagón/metabolismo , Hiperglucemia/patología , Obesidad/patología , Ubiquitina Tiolesterasa/metabolismo , Animales , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Retículo Endoplásmico/patología , Técnicas de Silenciamiento del Gen , Gluconeogénesis/fisiología , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ubiquitina Tiolesterasa/genética
3.
Cancer Cell Int ; 21(1): 390, 2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34289837

RESUMEN

BACKGROUND: miR-198 is involved in the formation, migration, invasion, and metastasis of various malignant cancers. However, the function and mechanism of action of miR-198 in the tumorigenesis of renal cell carcinoma (RCC) remain elusive. Here, we aimed to explore the role of miR198 in RCC. METHODS: Immunohistochemistry was performed to estimate the level of survivin in RCC sections. Quantitative real-time polymerase chain reaction was performed to determine the expression level of miR-198 in fresh RCC tissues. Furthermore, the target relationship between miR-198 and BIRC5 was predicted using the TargetScanHuman 7.2 database and verified via dual-luciferase reporter assay and western blotting. The effects of miR-198 on the viability, apoptosis, invasion, and migration of A498 and ACHN cells were studied using Cell Counting Kit-8, flow cytometry, transwell migration assay, and wound healing assay, respectively. Additionally, a xenograft nude mouse model was established to evaluate the effect of miR-198 on RCC tumorigenesis. RESULTS: The expression levels of BIRC5 and miR-198 were respectively higher and lower in RCC tissues than those in normal adjacent tissues. Furthermore, miR-198 could inhibit luciferase activity and reduce the protein level of survivin without affecting the BIRC5 mRNA levels. miR-198 inhibited cell viability, migration, and invasion and promoted cell apoptosis; co-transfection with BIRC5 could rescue these effects. Moreover, miR-198 could repress tumor growth in the xenograft nude mouse model of RCC. CONCLUSIONS: Our study demonstrates that miR-198 suppresses RCC progression by targeting BIRC5.

4.
Acta Pharmacol Sin ; 41(9): 1246-1254, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32210356

RESUMEN

Mitophagy is a degradative pathway that mediates the degradation of the entire mitochondria, and defects in this process are implicated in many diseases including cancer. In mammals, mitophagy is mediated by BNIP3L (also known as NIX) that is a dual regulator of mitochondrial turnover and programmed cell death pathways. Acute myeloid leukemia (AML) cells with deficiency of BNIP3L are more sensitive to mitochondria-targeting drugs. But small molecular inhibitors for BNIP3L are currently not available. Some immunomodulatory drugs (IMiDs) have been proved by FDA for hematologic malignancies, however, the underlining molecular mechanisms are still elusive, which hindered the applications of BNIP3L inhibition for AML treatment. In this study we carried out MS-based quantitative proteomics analysis to identify the potential neosubstrates of a novel thalidomide derivative CC-885 in A549 cells. In total, we quantified 5029 proteins with 36 downregulated in CRBN+/+ cell after CC-885 administration. Bioinformatic analysis showed that macromitophagy pathway was enriched in the negative pathway after CC-885 treatment. We further found that CC-885 caused both dose- and time-dependent degradation of BNIP3L in CRBN+/+, but not CRBN-/- cell. Thus, our data uncover a novel role of CC-885 in the regulation of mitophagy by targeting BNIP3L for CRL4CRBN E3 ligase-dependent ubiquitination and degradation, suggesting that CC-885 could be used as a selective BNIP3L degradator for the further investigation. Furthermore, we demonstrated that CC-885 could enhance AML cell sensitivity to the mitochondria-targeting drug rotenone, suggesting that combining CC-885 and mitochondria-targeting drugs may be a therapeutic strategy for AML patients.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Mitofagia/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Talidomida/análogos & derivados , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Células HEK293 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Rotenona/farmacología , Talidomida/farmacología , Ubiquitinación/efectos de los fármacos
5.
Biochim Biophys Acta ; 1853(8): 1808-17, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25940838

RESUMEN

The differentiation from constantly dividing epithelial cells into secondary fiber cells is a key step during lens development. Failure in this process, which requires cell proliferation inhibition and cell cycle exit, causes cataract formation. HSF4 (Heat Shock Transcription Factor 4) gene mutations may lead to both congenital and senile cataract. However, how HSF4 mutations induce cataract formation remains obscure. In this study, we demonstrate that HSF4 can suppress the proliferation of human lens epithelial cells (HLECs) by promoting G1/S arrest in a p53-dependent manner. In contrast, HSF4 with cataract causative mutations fail to cause cell cycle arrest and have no obvious effect on cell proliferation. We further identify that HSF4 recruits p53 in the nucleus and promotes its transcriptional activity, leading to the expression of its target gene p21 in HLECs. HSF4, but not its cataract-causing mutants, stabilizes p53 protein and inhibits its ubiquitin degradation. Our data reveal that HSF4 may work as a switch between lens epithelial cell proliferation and secondary fiber cell differentiation, a process which mainly depends on p53. Through demonstration of this novel downstream pathway of HSF4, our results help uncover the pathogenic mechanisms caused by HSF4 mutations.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Epiteliales/fisiología , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Cristalino , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Genes de Cambio , Factores de Transcripción del Choque Térmico , Humanos , Cristalino/citología , Cristalino/fisiología , Proteínas Mutantes/metabolismo , Unión Proteica , Estabilidad Proteica , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Células Tumorales Cultivadas
6.
Biochim Biophys Acta ; 1832(8): 1167-72, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23507146

RESUMEN

HSF4 mutations lead to both congenital and age-related cataract. The purpose of this study was to explore the mechanism of cataract formation caused by HSF4 mutations. The degradation of nuclear DNA is essential for the lens fiber differentiation. DNase 2ß (DLAD) is highly expressed in lens cells, and mice with deficiencies in the DLAD gene develop nuclear cataracts. In this study, we found that HSF4 promoted the expression and DNase activity of DLAD by directly binding to the DLAD promoter. In contrast, HSF4 cataract causative mutations failed to bind to the DLAD promoter, abrogating the expression and DNase activity of DLAD. These results were confirmed by HSF4 knockdown in zebrafish, which led to incomplete de-nucleation of the lens and decreased expression and activity of DLAD. Together, our results suggest that HSF4 exerts its function on lens differentiation via positive regulation of DLAD expression and activity, thus facilitating de-nucleation of lens fiber cells. Our demonstration that HSF4 cataract causative mutations abrogate the induction of DLAD expression reveals a novel molecular mechanism regarding how HSF4 mutations cause cataractogenesis.


Asunto(s)
Catarata/fisiopatología , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/biosíntesis , Cristalino/fisiología , Factores de Transcripción/metabolismo , Animales , Catarata/genética , Catarata/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/fisiología , Células Cultivadas , ADN/genética , Proteínas de Unión al ADN/genética , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Factores de Transcripción del Choque Térmico , Humanos , Cristalino/metabolismo , Mutación , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Pez Cebra
7.
Biochim Biophys Acta ; 1822(8): 1308-15, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22587838

RESUMEN

Heat shock factor protein 4 (HSF4) is expressed exclusively in the ocular lens and plays a critical role in the lens formation and differentiation. Mutations in the HSF4 gene lead to congenital and senile cataract. However, the molecular mechanisms causing this disease have not been well characterized. DNA damage in lens is a crucial risk factor in senile cataract formation, and its timely repair is essential for maintaining the lens' transparency. Our study firstly found evidence that HSF4 contributes to the repair of DNA strand breaks. Yet, this does not occur with cataract causative mutations in HSF4. We verify that DNA damage repair is mediated by the binding of HSF4 to a heat shock element in the Rad51 promoter, a gene which assists in the homologous recombination (HR) repair of DNA strand breaks. HSF4 up-regulates Rad51 expression while mutations in HSF4 fail, and DNA does not get repaired. Camptothecin, which interrupts the regulation of Rad51 by HSF4, also affects DNA damage repair. Additionally, with HSF4 knockdown in the lens of Zebrafish, DNA damage was observed and the protein level of Rad51 was significantly lower. Our study presents the first evidence demonstrating that HSF4 plays a role in DNA damage repair and may contribute a better understanding of congenital cataract formation.


Asunto(s)
Catarata/genética , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Choque Térmico/genética , Cristalino/metabolismo , Recombinasa Rad51/metabolismo , Factores de Transcripción/metabolismo , Animales , Camptotecina/farmacología , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/metabolismo , Humanos , Regiones Promotoras Genéticas , Recombinasa Rad51/genética , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Transfección , Pez Cebra
8.
Front Oncol ; 13: 1077640, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36969058

RESUMEN

Background: Glioblastoma multiforme (GBM) is the most lethal brain cancer in adults, characterized by rapid growth, extensive invasiveness, and poor prognosis, and there is still a lack of effective treatments. Here, we aimed to explore the role of triptolide (TPL), purified from Tripterygium wilfordii Hook F, on glioblastoma cell growth, apoptosis, proliferation, migration and invasion, as well as potential underlying mechanisms. Methods: The publicly available clinical data of Brain Lower Grade Glioma (LGG) from The Cancer Genome Atlas (TCGA) had been screened to observe PROX1 expression. The Kaplan-Meier analysis was used to analyze the relationship between PROX1 expression and GBM prognosis. CCK8, cell cycle, EDU, apoptosis, wound healing, and transwell assays were performed to detect the effects of TPL on glioblastoma U251 cell viability, cell cycle, proliferation, apoptosis, migration and invasion, respectively. Further, a soft agar colony assay was used to calculate the growth of glioblastoma cells. The qRT-PCR and western blot were conducted to quantify PROX1 mRNA and protein levels. The transcriptional regulation of TPL was detected by Dual luciferase reporter assay. Results: We found that TPL inhibited glioblastoma cell viability, proliferation, cell cycle, migration and invasion, but enhanced apoptosis in a dose-dependent manner. The expression of cell cycle inhibitor, P21, and pro-apoptosis factor, Bax was increased, while invasion-related factors MMP2 and MMP9 were silenced after TPL treatments. Mechanistically, TPL showed transcriptional inhibition of PROX1 appearance. Moreover, ectopic expression of PROX1 partially rescued the effects of TPL on glioblastoma cell viability, proliferation, apoptosis, migration and invasion, and on the expression of cell function-related genes. Conclusion: This study verified that TPL inhibited the progression of glioblastoma cells by transcriptionally depressing the expression of PROX1.

9.
Gene ; 655: 30-34, 2018 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-29454088

RESUMEN

The major causes for cataract formation are free radicals, which are neutralized by the endogenous antioxidants. However, how the human lens clean these harmful free radicals is still unclear. Transcriptional factor heat shock factor 4 (HSF4) is a cataract-causing gene and plays important roles during lens development. Here we show that HMOX-1, an anti-oxidase, is a bona fide transcriptional target gene of HSF4 in HLECs (human lens epithelial cells). HSF4 directly binds to the HSE element in HMOX-1 promoter to mediate its mRNA transcription and protein accumulation. The HSE element located at the region of -389 bp to -362 bp upstream from the TSS (transcription start site), which is critical for HMOX-1 transcriptional activation. Furthermore, knockdown of HSF4 by siRNA inhibited HMOX-1 expression. Thus, these data revealed a novel transcription target of HSF4 and provided new insights into anti-oxidation regulation in lens and age-related cataract.


Asunto(s)
Células Epiteliales/metabolismo , Factores de Transcripción del Choque Térmico/fisiología , Hemo-Oxigenasa 1/genética , Cristalino/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Factores de Transcripción del Choque Térmico/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Cristalino/citología , Regiones Promotoras Genéticas , Conejos , Elementos de Respuesta , Activación Transcripcional
10.
Diabetes ; 67(11): 2227-2238, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30181160

RESUMEN

Obesity is usually associated with an increased risk of nonalcoholic fatty liver disease that is characterized by accumulation of excessive triglyceride (TG) in hepatocytes. However, the factors involved in the obesity-induced hepatosteatosis are poorly defined. Here, we report that SRY-box containing gene 4 (Sox4), a transcription factor that regulates cell proliferation and differentiation, plays an important role in hepatic TG metabolism. Sox4 expression levels are markedly upregulated in livers of obese rodents and humans. Adenovirus-medicated overexpression of Sox4 in the livers of lean mice promotes liver steatosis, whereas liver-specific knockdown of Sox4 ameliorates TG accumulation and improves insulin resistance in obese mice. At the molecular level, we show that Sox4 could directly control the transcription of SREBP-1c gene through binding to its proximal promoter region. Thus, we have identified Sox4 as an important component of hepatic TG metabolism.


Asunto(s)
Hígado Graso/metabolismo , Hígado/metabolismo , Obesidad/metabolismo , Factores de Transcripción SOXC/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo , Animales , Hígado Graso/genética , Hígado Graso/patología , Regulación de la Expresión Génica , Humanos , Resistencia a la Insulina/fisiología , Hígado/patología , Masculino , Ratones , Ratones Obesos , Obesidad/genética , Obesidad/patología , Regiones Promotoras Genéticas , Factores de Transcripción SOXC/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Regulación hacia Arriba
11.
Nat Commun ; 9(1): 4770, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30425250

RESUMEN

Ubiquitin-specific protease 14 (USP14) is one of the major proteasome-associated deubiquitinating enzymes critical for proteome homeostasis. However, substrates of USP14 remain largely unknown, hindering the understanding of its functional roles. Here we conduct a comprehensive proteome, ubiquitinome and interactome analysis for USP14 substrate screening. Bioinformatics analysis reveals broad new potential roles of USP14, especially in lipid and carbohydrate metabolism. Among the potential substrates identified, we show that fatty acid synthase (FASN), a key enzyme involved in hepatic lipogenesis, is a bona fide substrate of USP14. USP14 directly interacts with and increases FASN stability. As a result, overexpression of USP14 promotes liver triglyceride accumulation in C57BL/6 mice, whereas genetic ablation or pharmacological inhibition of USP14 ameliorates hepatosteatosis, hyperglycemia and insulin resistance in obese mice. In conclusion, our findings reveal for the first time an indispensable role of USP14 in hepatosteatosis through FASN stabilization.


Asunto(s)
Ácido Graso Sintasas/metabolismo , Proteoma , Ubiquitina Tiolesterasa/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Biología Computacional , Técnicas de Silenciamiento del Gen , Homeostasis , Humanos , Hiperglucemia , Resistencia a la Insulina , Lipogénesis , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Triglicéridos/análisis , Ubiquitina Tiolesterasa/genética , Regulación hacia Arriba
12.
Diabetes ; 66(3): 689-698, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27932386

RESUMEN

Insulin resistance is a critical factor in the development of metabolic disorders, including type 2 diabetes (T2DM). However, its molecular mechanisms remain incompletely understood. In this study, we found that F-box only protein 2 (FBXO2), a substrate recognition component of the Skp1-Cul1-F-box protein (SCF) E3 ubiquitin ligase complex, was upregulated in livers of obese mice. Furthermore, using a protein purification approach combined with high-performance liquid chromatography/tandem mass spectrometry, we carried out a system-wide screening of FBXO2 substrates, in which the insulin receptor (IR) was identified as a substrate for FBXO2. SCFFBXO2 acts as an E3 ligase targeting the IR for ubiquitin-dependent degradation to regulate insulin signaling integrity. As a result, adenovirus-mediated overexpression of FBXO2 in healthy mice led to hyperglycemia, glucose intolerance, and insulin resistance, whereas ablation of FBXO2 alleviated diabetic phenotypes in obese mice. Therefore, our results identify SCFFBXO2 as an E3 ligase for the IR in the liver, which might provide a novel therapeutic target for treating T2DM and related metabolic disorders.


Asunto(s)
Glucemia/metabolismo , Proteínas F-Box/metabolismo , Resistencia a la Insulina , Hígado/metabolismo , Obesidad/metabolismo , Receptor de Insulina/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Cromatografía Líquida de Alta Presión , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Células HEK293 , Homeostasis , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Obesos , Espectrometría de Masas en Tándem
13.
Oncotarget ; 8(57): 97178-97186, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29228602

RESUMEN

Mof4 family associated protein 1 (MRFAP1) is a 14 kDa nuclear protein, which involves in maintaining normal histone modification levels by negatively regulating recruitment of the NuA4 (nucleosome acetyltransferase of H4) histone acetyltransferase complex to chromatin. MRFAP1 has been identified as one of the most up-regulated proteins after NEDD8 (neural precursor cell expressed developmentally down-regulated 8) inhibition in multiple human cell lines. However, the biological function of MRFAP1 and the E3 ligase that targets MRFAP1 for destruction remain mysterious. Here we show, by using an immunoprecipitation-based proteomics screen, that MRFAP1 is an interactor of the F-box protein FBXW8. MRFAP1 is degraded by means of the ubiquitin ligase Cul7/FBXW8 during mitotic anaphase-telophase transition and accumulated in mitotic metaphase. Overexpression of FBXW8 increased the polyubiquitination and decreased the stability of MRFAP1, whereas knockdown of FBXW8 prolonged the half-life of MRFAP1. Moreover, forced expression of MRFAP1 in HeLa cells caused growth retardation and genomic instability, leading to severe mitotic cell death. Thus, Cul7/FBXW8-mediated destruction of MRFAP1 is a regulatory component monitoring the anaphase-telophase transition and preventing genomic instability.

15.
J Invest Dermatol ; 133(9): 2221-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23519333

RESUMEN

Dyschromatosis universalis hereditaria (DUH) is a pigmentary genodermatosis characterized by a mixture of hyperpigmented and hypopigmented macules distributed randomly over the body. No causative genes have been reported to date. In this study, we investigated a large five-generation Chinese family with DUH. After excluding the two known DUH loci, we performed genome-wide linkage analysis and identified a DUH locus on chromosome 2q33.3-q36.1 with a maximum LOD score of 3.49 with marker D2S2382. Exome sequencing identified a c.1067T>C (p.Leu356Pro) mutation in exon 3 of ABCB6 (ATP-binding cassette subfamily B, member 6) in the DUH family. Two additional missense mutations, c.508A>G (p.Ser170Gly) in exon 1 and c.1736G>A (p.Gly579Glu) in exon 12 of ABCB6, were found in two out of six patients by mutational screening using sporadic DUH patients. Immunohistologic examination in biopsy specimens showed that ABCB6 is expressed in the epidermis and had a diffuse cytoplasmic distribution. Examination of subcellular localization of wild-type ABCB6 in a B16 mouse melanoma cell line revealed that it is localized to the endosome-like compartment and dendrite tips, whereas disease-causing mutations of ABCB6 resulted in its retention in the Golgi apparatus. Our studies identified ABCB6 as the first pathogenic gene associated with DUH. These findings suggest that ABCB6 may be a physiological factor for skin pigmentation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Pueblo Asiatico/genética , Cromosomas Humanos Par 2 , Trastornos de la Pigmentación/congénito , Enfermedades Cutáneas Genéticas/genética , Pigmentación de la Piel/genética , Secuencia de Aminoácidos , Niño , Femenino , Genes Dominantes , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Trastornos de la Pigmentación/genética , Trastornos de la Pigmentación/patología , Homología de Secuencia de Aminoácido , Piel/patología , Enfermedades Cutáneas Genéticas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA