Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.159
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-37511054

RESUMO

Skin pigmentation ensures efficient photoprotection and relies on the pigment melanin, which is produced by epidermal melanocytes and transferred to surrounding keratinocytes. While the molecular mechanisms of melanin synthesis and transport in melanocytes are now well characterized, much less is known about melanin transfer and processing within keratinocytes. Over the past few decades, distinct models have been proposed to explain how melanin transfer occurs at the cellular and molecular levels. However, this remains a debated topic, as up to four different models have been proposed, with evidence presented supporting each. Here, we review the current knowledge on the regulation of melanin exocytosis, internalization, processing, and polarization. Regarding the different transfer models, we discuss how these might co-exist to regulate skin pigmentation under different conditions, i.e., constitutive and facultative skin pigmentation or physiological and pathological conditions. Moreover, we discuss recent evidence that sheds light on the regulation of melanin exocytosis by melanocytes and internalization by keratinocytes, as well as how melanin is stored within these cells in a compartment that we propose be named the melanokerasome. Finally, we review the state of the art on the molecular mechanisms that lead to melanokerasome positioning above the nuclei of keratinocytes, forming supranuclear caps that shield the nuclear DNA from UV radiation. Thus, we provide a comprehensive overview of the current knowledge on the molecular mechanisms regulating skin pigmentation, from melanin exocytosis by melanocytes and internalization by keratinocytes to processing and polarization within keratinocytes. A better knowledge of these molecular mechanisms will clarify long-lasting questions in the field that are crucial for the understanding of skin pigmentation and can shed light on fundamental aspects of organelle biology. Ultimately, this knowledge can lead to novel therapeutic strategies to treat hypo- or hyper-pigmentation disorders, which have a high socio-economic burden on patients and healthcare systems worldwide, as well as cosmetic applications.


Assuntos
Melaninas , Melanócitos , Humanos , Melanócitos/fisiologia , Queratinócitos/fisiologia , Epiderme , Pigmentação da Pele , Melanossomas
2.
Front Immunol ; 14: 1294919, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38239366

RESUMO

Vitiligo is an autoimmune disease that leads to disfiguring depigmented lesions of skin and mucosa. Although effective treatments are available for vitiligo, there are still some patients with poor responses to conventional treatment. Refractory vitiligo lesions are mostly located on exposed sites such as acral sites and lips, leading to significant life stress. Understanding the causes of refractory vitiligo and developing targeted treatments are essential to enhance vitiligo outcomes. In this review, we summarized recent treatment approaches and some potential methods for refractory vitiligo. Janus kinase inhibitors have shown efficacy in refractory vitiligo. A variety of surgical interventions and fractional carbon dioxide laser have been widely applied to combination therapies. Furthermore, melanocyte regeneration and activation therapies are potentially effective strategies. Patients with refractory vitiligo should be referred to psychological monitoring and interventions to reduce the potential pathogenic effects of chronic stress. Finally, methods for depigmentation and camouflage may be beneficial in achieving uniform skin color and improved quality of life. Our ultimate focus is to provide alternative options for refractory vitiligo and to bring inspiration to future research.


Assuntos
Vitiligo , Humanos , Vitiligo/terapia , Qualidade de Vida , Resultado do Tratamento , Terapia Combinada , Melanócitos/fisiologia
3.
Commun Biol ; 5(1): 17, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017622

RESUMO

Countless biophysical studies have sought distinct markers in the cellular mechanical response that could be linked to morphogenesis, homeostasis, and disease. Here, an iterative-fitting methodology visualizes the time-dependent viscoelastic behavior of human skin cells under physiologically relevant conditions. Past investigations often involved parameterizing elastic relationships and assuming purely Hertzian contact mechanics, which fails to properly account for the rich temporal information available. We demonstrate the performance superiority of the proposed iterative viscoelastic characterization method over standard open-search approaches. Our viscoelastic measurements revealed that 2D adherent metastatic melanoma cells exhibit reduced elasticity compared to their normal counterparts-melanocytes and fibroblasts, and are significantly less viscous than fibroblasts over timescales spanning three orders of magnitude. The measured loss angle indicates clear differential viscoelastic responses across multiple timescales between the measured cells. This method provides insight into the complex viscoelastic behavior of metastatic melanoma cells relevant to better understanding cancer metastasis and aggression.


Assuntos
Elasticidade/fisiologia , Fibroblastos/fisiologia , Melanócitos/fisiologia , Pele/citologia , Linhagem Celular Tumoral , Células Cultivadas , Fibroblastos/citologia , Humanos , Melanócitos/citologia , Melanoma/fisiopatologia , Neoplasias Cutâneas/fisiopatologia , Viscosidade
4.
Cell Rep ; 38(2): 110234, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021087

RESUMO

Melanocytes, the pigment-producing cells, are replenished from multiple stem cell niches in adult tissue. Although pigmentation traits are known risk factors for melanoma, we know little about melanocyte stem cell (McSC) populations other than hair follicle McSCs and lack key lineage markers with which to identify McSCs and study their function. Here we find that Tfap2b and a select set of target genes specify an McSC population at the dorsal root ganglia in zebrafish. Functionally, Tfap2b is required for only a few late-stage embryonic melanocytes, and is essential for McSC-dependent melanocyte regeneration. Fate mapping data reveal that tfap2b+ McSCs have multifate potential, and are the cells of origin for large patches of adult melanocytes, two other pigment cell types (iridophores and xanthophores), and nerve-associated cells. Hence, Tfap2b confers McSC identity in early development, distinguishing McSCs from other neural crest and pigment cell lineages, and retains multifate potential in the adult zebrafish.


Assuntos
Melanócitos/metabolismo , Células-Tronco/classificação , Fator de Transcrição AP-2/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Melanócitos/fisiologia , Pigmentação/genética , Pele/metabolismo , Pigmentação da Pele/genética , Células-Tronco/metabolismo , Fator de Transcrição AP-2/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
5.
J Med Genet ; 59(2): 105-114, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34667088

RESUMO

SOX10 belongs to a family of 20 SRY (sex-determining region Y)-related high mobility group box-containing (SOX) proteins, most of which contribute to cell type specification and differentiation of various lineages. The first clue that SOX10 is essential for development, especially in the neural crest, came with the discovery that heterozygous mutations occurring within and around SOX10 cause Waardenburg syndrome type 4. Since then, heterozygous mutations have been reported in Waardenburg syndrome type 2 (Waardenburg syndrome type without Hirschsprung disease), PCWH or PCW (peripheral demyelinating neuropathy, central dysmyelination, Waardenburg syndrome, with or without Hirschsprung disease), intestinal manifestations beyond Hirschsprung (ie, chronic intestinal pseudo-obstruction), Kallmann syndrome and cancer. All of these diseases are consistent with the regulatory role of SOX10 in various neural crest derivatives (melanocytes, the enteric nervous system, Schwann cells and olfactory ensheathing cells) and extraneural crest tissues (inner ear, oligodendrocytes). The recent evolution of medical practice in constitutional genetics has led to the identification of SOX10 variants in atypical contexts, such as isolated hearing loss or neurodevelopmental disorders, making them more difficult to classify in the absence of both a typical phenotype and specific expertise. Here, we report novel mutations and review those that have already been published and their functional consequences, along with current understanding of SOX10 function in the affected cell types identified through in vivo and in vitro models. We also discuss research options to increase our understanding of the origin of the observed phenotypic variability and improve the diagnosis and medical care of affected patients.


Assuntos
Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/fisiologia , Animais , Sistema Nervoso Entérico/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Perda Auditiva/genética , Doença de Hirschsprung/genética , Humanos , Síndrome de Kallmann/genética , Melanócitos/fisiologia , Mutação , Neoplasias/genética , Crista Neural/embriologia , Crista Neural/fisiologia , Fenótipo , Síndrome de Waardenburg/genética
6.
Clin Exp Dermatol ; 47(2): 251-258, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33999447

RESUMO

There is an increasing recognition of ethnic dermatology to reflect the increase in skin of colour (SOC) populations in the UK. Hyperpigmentary disorder is one of the commonest skin concerns in SOC but there has been limited training available in this field of dermatology. Variations in skin colour are genetically determined by the amount of melanin content, the eumelanin/pheomelanin ratio and the size of melanosomes, but is also influenced by other factors such as hormones and extrinsic factors such as ultraviolet radiation. Hyperpigmentation is a broad term to describe increased pigmentation in the skin, and making a correct diagnosis is an important first step in the successful management of hyperpigmentary disorders. A systematic approach based on the disease pathogenesis (e.g. reactive vs. nonreactive, increased melanin vs. increased number of cells or epidermal vs. dermal pigmentation) aided by a detailed history and clinical examination is the best way to diagnose a hyperpigmentary disorder. Based on its pathogenesis, management can be planned. For epidermal hyperpigmentation caused by increased melanin, topical skin-lightening agents targeting inhibition of tyrosinase or melanosome transfer and promotion of keratinocyte turnover can be used. Hydroquinone-containing cream is the gold-standard treatment for epidermal hyperpigmentation. Alternative treatments include laser toning or chemical peels. However, increased dermal pigmentation is more challenging to target with topical treatments. If hyperpigmentation is due to increased numbers of melanocytes or keratinocytes, high-fluence laser is the most appropriate treatment method.


Assuntos
Hiperpigmentação/diagnóstico , Diagnóstico Diferencial , Humanos , Hidroquinonas/administração & dosagem , Hiperpigmentação/etiologia , Hiperpigmentação/fisiopatologia , Hiperpigmentação/terapia , Melaninas/fisiologia , Melanócitos/fisiologia , Creme para a Pele
7.
J Invest Dermatol ; 142(1): 201-211, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34265328

RESUMO

Drug resistance mechanisms still characterize metastatic melanoma, despite the new treatments that have been recently developed. Targeting of the cGMP/protein kinase G pathway is emerging as a therapeutic approach in cancer research. In this study, we evaluated the anticancer effects of two polymeric-linked dimeric cGMP analogs able to bind and activate protein kinase G, called protein kinase G activators (PAs) 4 and 5. PA5 was identified as the most effective compound on melanoma cell lines as well as on patient-derived metastatic melanoma cells cultured as three-dimensional spheroids and in a zebrafish melanoma model. PA5 was able to significantly reduce cell viability, size, and invasion of melanoma spheroids. Importantly, PA5 showed a tumor-specific outcome because no toxic effect was observed in healthy melanocytes exposed to the cGMP analog. We defined that by triggering protein kinase G, PA5 interfered with the EGF pathway as shown by lower EGFR phosphorylation and reduction of activated, phosphorylated forms of protein kinase B and extracellular signal‒regulated kinase 1/2 in melanoma cells. Finally, PA5 significantly reduced the metastatic process in zebrafish. These studies open future perspectives for the cGMP analog PA5 as a potential therapeutic strategy for melanoma.


Assuntos
Antineoplásicos/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Melanócitos/fisiologia , Melanoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , GMP Cíclico/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Fosforilação , Transdução de Sinais , Peixe-Zebra
8.
Pigment Cell Melanoma Res ; 35(1): 38-51, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34467641

RESUMO

We previously described a novel in vitro culture technique for dedifferentiated human adult skin melanocytes. Melanocytes cultured in a defined, cholera toxin and PMA free medium became bipolar, unpigmented, and highly proliferative. Furthermore, TRP-1 and c-Kit expression disappeared and EGFR receptor and nestin expression were induced in the cells. Here, we further characterized the phenotype of these dedifferentiated cells and by comparing them to mature pigmented melanocytes we detected crucial steps in their phenotype change. Our data suggest that normal adult melanocytes easily dedifferentiate into pluripotent stem cells given the right environment. This dedifferentiation process described here for normal melanocyte is very similar to what has been described for melanoma cells, indicating that phenotype switching driven by environmental factors is a general characteristic of melanocytes that can occur independent of malignant transformation.


Assuntos
Desdiferenciação Celular , Plasticidade Celular , Melanócitos/fisiologia , Pele/citologia , Adulto , Proliferação de Células , Células Cultivadas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Melaninas/metabolismo , Melanócitos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Nestina/genética , Nestina/metabolismo , Oxirredutases/genética , Oxirredutases/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA-Seq , Transdução de Sinais , Transcriptoma , Adulto Jovem
9.
Elife ; 102021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34812139

RESUMO

Benign melanocytic nevi frequently emerge when an acquired BRAFV600E mutation triggers unchecked proliferation and subsequent arrest in melanocytes. Recent observations have challenged the role of oncogene-induced senescence in melanocytic nevus formation, necessitating investigations into alternative mechanisms for the establishment and maintenance of proliferation arrest in nevi. We compared the transcriptomes of melanocytes from healthy human skin, nevi, and melanomas arising from nevi and identified a set of microRNAs as highly expressed nevus-enriched transcripts. Two of these microRNAs-MIR211-5p and MIR328-3p-induced mitotic failure, genome duplication, and proliferation arrest in human melanocytes through convergent targeting of AURKB. We demonstrate that BRAFV600E induces a similar proliferation arrest in primary human melanocytes that is both reversible and conditional. Specifically, BRAFV600E expression stimulates either arrest or proliferation depending on the differentiation state of the melanocyte. We report genome duplication in human melanocytic nevi, reciprocal expression of AURKB and microRNAs in nevi and melanomas, and rescue of arrested human nevus cells with AURKB expression. Taken together, our data describe an alternative molecular mechanism for melanocytic nevus formation that is congruent with both experimental and clinical observations.


Lots of people have small dark patches on their skin known as moles. Most moles form when individual cells known as melanocytes in the skin acquire a specific genetic mutation in a gene called BRAF. This mutation causes the cells to divide rapidly to form the mole. After a while, most moles stop growing and remain harmless for the rest of a person's life. Melanoma is a type of skin cancer that develops from damaged melanocytes. The same mutation in BRAF that is found in moles is also present in half of all cases of melanoma. Unlike in moles, the melanoma-causing mutation makes the melanocytes divide rapidly to form a tumor that keeps on growing indefinitely. It remains unclear why the same genetic mutation in the BRAF gene has such different consequences in moles and melanomas. To address this question, McNeal et al. used genetic approaches to study melanocytes from moles and melanomas. The experiments identified some molecules known as microRNAs that are present at higher levels in moles than in melanomas. Increasing the levels of two of these microRNAs in melanocytes from human skin stopped the cells from growing and dividing by inhibiting a gene called AURKB. This suggested that these microRNAs are responsible for halting the growth of moles. Introducing the mutated form of BRAF into melanocytes also stopped cells from growing and dividing by inhibiting AURKB. However, changing the environment surrounding the cells reversed this effect and allowed the melanocytes to resume dividing. In this way the mutated form of BRAF acts like a switch that allows melanocytes in skin cancers to start growing again under certain conditions. Further experiments found that a drug called barasertib is able to inhibit the growth of melanoma cells with the mutant form of BRAF. Future work will investigate whether it is possible to use this drug and other tools to stop skin cancer tumors from growing, and possibly even prevent skin tumors from forming in the first place.


Assuntos
Aurora Quinase B/genética , Melanócitos/fisiologia , MicroRNAs/metabolismo , Mitose/genética , Proteínas Proto-Oncogênicas B-raf/genética , Aurora Quinase B/metabolismo , Humanos , Proteínas Proto-Oncogênicas B-raf/metabolismo , Transdução de Sinais
10.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34360837

RESUMO

Skin pigmentation can occur due to increased melanin, including melanocyte proliferation, melanin biosynthesis, or melanocyte migration. There are many factors that influence the melanin production process, but the role of neurotransmitters in this process is still unclear. We found that histamine and serotonin influence the different stages of melanogenesis and melanogenesis, which increase melanogenesis. Since then, several related papers have been published, and from these papers, it has been recognised that the role of neurotransmitters in skin-pigment-related diseases needs to be summarised. By introducing the role of neurotransmitters in the regulation of various pigment disorders, including vitiligo and melasma, through this review, many researchers can be expected to try to apply neurotransmitter-related agonists and antagonists as treatments for skin pigment disorders.


Assuntos
Neurotransmissores/metabolismo , Transtornos da Pigmentação/metabolismo , Receptores de Neurotransmissores/metabolismo , Pigmentação da Pele , Animais , Humanos , Melaninas/metabolismo , Melanócitos/metabolismo , Melanócitos/fisiologia , Melanose , Neurotransmissores/fisiologia , Transtornos da Pigmentação/fisiopatologia , Receptores de Neurotransmissores/fisiologia , Vitiligo
11.
Cell Mol Life Sci ; 78(16): 6033-6049, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34274976

RESUMO

Melanocytes are pigmented cells residing mostly in the skin and hair follicles of vertebrates, where they contribute to colouration and protection against UV-B radiation. However, the spectrum of their functions reaches far beyond that. For instance, these pigment-producing cells are found inside the inner ear, where they contribute to the hearing function, and in the heart, where they are involved in the electrical conductivity and support the stiffness of cardiac valves. The embryonic origin of such extracutaneous melanocytes is not clear. We took advantage of lineage-tracing experiments combined with 3D visualizations and gene knockout strategies to address this long-standing question. We revealed that Schwann cell precursors are recruited from the local innervation during embryonic development and give rise to extracutaneous melanocytes in the heart, brain meninges, inner ear, and other locations. In embryos with a knockout of the EdnrB receptor, a condition imitating Waardenburg syndrome, we observed only nerve-associated melanoblasts, which failed to detach from the nerves and to enter the inner ear. Finally, we looked into the evolutionary aspects of extracutaneous melanocytes and found that pigment cells are associated mainly with nerves and blood vessels in amphibians and fish. This new knowledge of the nerve-dependent origin of extracutaneous pigment cells might be directly relevant to the formation of extracutaneous melanoma in humans.


Assuntos
Encéfalo/fisiologia , Orelha Interna/fisiologia , Coração/fisiologia , Meninges/fisiologia , Sistema Nervoso/fisiopatologia , Células de Schwann/fisiologia , Anfíbios/metabolismo , Anfíbios/fisiologia , Animais , Encéfalo/metabolismo , Linhagem da Célula/fisiologia , Orelha Interna/metabolismo , Desenvolvimento Embrionário/fisiologia , Feminino , Peixes/metabolismo , Peixes/fisiologia , Melanócitos/metabolismo , Melanócitos/fisiologia , Meninges/metabolismo , Camundongos , Sistema Nervoso/metabolismo , Gravidez , Receptor de Endotelina B/metabolismo , Células de Schwann/metabolismo
12.
PLoS One ; 16(5): e0251121, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33983985

RESUMO

Several angiogenesis-dependent diseases, including age-related macular degeneration and infantile hemangioma, display differential prevalence among Black, as compared to White individuals. Although socioeconomic status and genetic architecture have been suggested as explaining these differences, we have recently shown that pigment production per se might be involved. For example, we have shown that the extracellular protein fibromodulin is a pro-angiogenic factor highly secreted by melanocytes in White but not Black individuals. Still, additional pigment-dependent angiogenic factors and their molecular mechanisms remain to be identified. Understanding the contribution of pigmentation to angiogenesis in health and disease is essential for precision medicine of angiogenesis-dependent diseases with racial disparity. Toward that goal, we compared the transcriptomes of Black and White individuals in three tissues with angiogenic activity, namely artery, whole blood, and skin. We identified several differentially expressed angiogenesis pathways, including artery morphogenesis, regulation of endothelial cell chemotaxis, and cellular response to vascular endothelial growth factor stimulus. We then demonstrated that the expression of key genes in these pathways is directly modulated by the degree of pigmentation. We further identified the precise pigment production pathway controlling the expression of these genes, namely melanocortin 1 receptor (MC1R) signaling. These results demonstrate pigment-mediated regulation of angiogenesis-related pathways and their driver genes across human tissues.


Assuntos
Melanócitos/metabolismo , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , Animais , Artérias/metabolismo , População Negra/genética , Sangue/metabolismo , Bases de Dados Genéticas , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Masculino , Melanócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/imunologia , Especificidade de Órgãos/genética , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 1 de Melanocortina/metabolismo , Pele/metabolismo , Transcriptoma/genética , População Branca/genética
13.
Front Immunol ; 12: 630892, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717163

RESUMO

Atopic dermatitis (AD) typically starts in infancy or early childhood, showing spontaneous remission in a subset of patients, while others develop lifelong disease. Despite an increased understanding of AD, factors guiding its natural course are only insufficiently elucidated. We thus performed suction blistering in skin of adult patients with stable, spontaneous remission from previous moderate-to-severe AD during childhood. Samples were compared to healthy controls without personal or familial history of atopy, and to chronic, active AD lesions. Skin cells and tissue fluid obtained were used for single-cell RNA sequencing and proteomic multiplex assays, respectively. We found overall cell composition and proteomic profiles of spontaneously healed AD to be comparable to healthy control skin, without upregulation of typical AD activity markers (e.g., IL13, S100As, and KRT16). Among all cell types in spontaneously healed AD, melanocytes harbored the largest numbers of differentially expressed genes in comparison to healthy controls, with upregulation of potentially anti-inflammatory markers such as PLA2G7. Conventional T-cells also showed increases in regulatory markers, and a general skewing toward a more Th1-like phenotype. By contrast, gene expression of regulatory T-cells and keratinocytes was essentially indistinguishable from healthy skin. Melanocytes and conventional T-cells might thus contribute a specific regulatory milieu in spontaneously healed AD skin.


Assuntos
Dermatite Atópica/imunologia , Melanócitos/fisiologia , Pele/imunologia , Linfócitos T/imunologia , Adulto , Dermatite Atópica/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteômica , Transcriptoma , Adulto Jovem
15.
Hum Cell ; 34(4): 1093-1102, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33768511

RESUMO

Perivascular-resident macrophage-like melanocytes (PVM/Ms) can upregulate the expression of tight junction-related proteins in endothelial cells (ECs) by secreting pigment epithelial-derived factor (PEDF), and thereby regulate the permeability of the intrastrial fluid-blood barrier critical for maintaining inner ear homeostasis. This study aimed to investigate the effects of long non-coding RNA (lncRNA) Rian on cell growth of PVM/Ms and PVM/Ms regulation of intrastrial fluid-blood barrier integrity mediated by PEDF. Rian was downregulated in the aged cochlea from 12-month-old C57BL/6 mice. Rian overexpression inhibited cell apoptosis and promoted cell viability of hypoxia-injured PVM/Ms as well as increased the concentration and expression of PEDF secreted by PVM/Ms. In contrast, Rian silencing exerted the opposite effects. Furthermore, in a cell co-culture model of ECs and PVM/Ms, Rian overexpression in PVM/Ms increased the expression of the junction-associated proteins in co-cultured ECs, and this effect was abrogated by blockade of PEDF by anti-PEDF in PVM/Ms. Further mechanistical investigation revealed that Rian promoted STAT3 nuclear translocation and activation by binding to FUS, and thereby promoted the secretion of PEDF. Collectively, Rian attenuates PVM/Ms injury and strengthens the ability of PVM/Ms to maintain the integrity of the endothelial barrier by promoting PEDF expression.


Assuntos
Células Endoteliais/metabolismo , Proteínas do Olho/metabolismo , Expressão Gênica/genética , Melanócitos/fisiologia , Fatores de Crescimento Neural/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , RNA Longo não Codificante/fisiologia , Serpinas/metabolismo , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Animais , Apoptose/genética , Sobrevivência Celular/genética , Células Cultivadas , Cóclea/metabolismo , Proteínas do Olho/genética , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/genética , Proteínas Nucleares/metabolismo , Ligação Proteica/genética , Proteína FUS de Ligação a RNA/metabolismo , Fator de Transcrição STAT3/metabolismo , Serpinas/genética
16.
Hematol Oncol Clin North Am ; 35(1): 29-56, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33759772

RESUMO

Melanoma skin cancer is derived from skin melanocytes and has a high risk of metastatic spread. The era of molecular genetics and next-generation sequencing has uncovered the role of oncogenic BRAFV600E mutations in many melanomas, validated the role of ultraviolet-induced DNA mutations in melanoma formation, and uncovered many of the molecular events that occur during melanoma development. Targeted therapies and immunotherapy have dramatically improved outcomes and provided an increased rate of cure for metastatic melanoma. This article reviews the formation of melanoma, the molecular events involved in melanoma growth and metastasis, and the biology underlying resistance to melanoma therapies.


Assuntos
Carcinogênese , Melanócitos , Melanoma , Fator de Transcrição Associado à Microftalmia , Neoplasias Cutâneas , Carcinogênese/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Imunoterapia , Melanócitos/patologia , Melanócitos/fisiologia , Melanoma/classificação , Melanoma/genética , Melanoma/fisiopatologia , Melanoma/terapia , Fator de Transcrição Associado à Microftalmia/fisiologia , Neoplasias Cutâneas/classificação , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/fisiopatologia , Neoplasias Cutâneas/terapia , Raios Ultravioleta/efeitos adversos
17.
Clin Exp Dermatol ; 46(4): 646-650, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33098692

RESUMO

In this review we show how the neuronal theory is relevant to the convergence theory for the mechanism causing vitiligo, especially the segmental type. Neuropeptides and neurotransmitters, such as neuropeptide Y and dopamine, can be central to the pathological mechanisms of melanocyte destruction. They link into a bidirectional network connecting cutaneous nerves, the neuroendocrine axis and the immune system, and through their local influence on cutaneous inflammation, to the antigen-specific regulatory T cells and the chemokine ligand type 9/chemokine receptor type 1 axis, which is thought to be the final pathway for melanocyte destruction.


Assuntos
Inibidores de Janus Quinases/uso terapêutico , Melanócitos/fisiologia , Neuropeptídeos/fisiologia , Vitiligo/fisiopatologia , Humanos , Janus Quinase 1/antagonistas & inibidores , Melanócitos/efeitos dos fármacos , Pele/inervação , Vitiligo/tratamento farmacológico , Vitiligo/genética , Vitiligo/imunologia
18.
Mol Biol Evol ; 38(3): 1122-1136, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33212507

RESUMO

Visible pigmentation phenotypes can be used to explore the regulation of gene expression and the evolution of coat color patterns in animals. Here, we performed whole-genome and RNA sequencing and applied genome-wide association study, comparative population genomics and biological experiments to show that the 2,809-bp-long LINE-1 insertion in the ASIP (agouti signaling protein) gene is the causative mutation for the white coat phenotype in swamp buffalo (Bubalus bubalis). This LINE-1 insertion (3' truncated and containing only 5' UTR) functions as a strong proximal promoter that leads to a 10-fold increase in the transcription of ASIP in white buffalo skin. The 165 bp of 5' UTR transcribed from the LINE-1 is spliced into the first coding exon of ASIP, resulting in a chimeric transcript. The increased expression of ASIP prevents melanocyte maturation, leading to the absence of pigment in white buffalo skin and hairs. Phylogenetic analyses indicate that the white buffalo-specific ASIP allele originated from a recent genetic transposition event in swamp buffalo. Interestingly, as a similar LINE-1 insertion has been identified in the cattle ASIP gene, we discuss the convergent mechanism of coat color evolution in the Bovini tribe.


Assuntos
Proteína Agouti Sinalizadora/genética , Evolução Biológica , Búfalos/genética , Elementos Nucleotídeos Longos e Dispersos , Pigmentação/genética , Proteína Agouti Sinalizadora/metabolismo , Animais , Búfalos/metabolismo , Bovinos , Elementos de DNA Transponíveis , Feminino , Masculino , Melanócitos/fisiologia , Fenótipo , Regiões Promotoras Genéticas , Pele/metabolismo , Sequenciamento Completo do Genoma
19.
Exp Dermatol ; 30(4): 560-571, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33320376

RESUMO

In the light of substantial discoveries in epithelial and hair pigmentation pathophysiology, this review summarizes the current understanding of skin pigmentation mechanisms. Melanocytes are pigment-producing cells, and their key regulating transcription factor is the melanocyte-specific microphthalmia-associated transcription factor (m-MITF). Ultraviolet (UV) radiation is a unique modulator of skin pigmentation influencing tanning pathways. The delayed tanning pathway occurs as UVB produces keratinocyte DNA damage, causing p53-mediated expression of the pro-opiomelanocortin (POMC) gene that is processed to release α-melanocyte-stimulating hormone (α-MSH). α-MSH stimulates the melanocortin 1 receptor (MC1R) on melanocytes, leading to m-MITF expression and melanogenesis. POMC cleavage also releases ß-endorphin, which creates a neuroendocrine pathway that promotes UV-seeking behaviours. Mutations along the tanning pathway can affect pigmentation and increase the risk of skin malignancies. MC1R variants have received considerable attention, yet the allele is highly polymorphic with varied phenotypes. Vitiligo presents with depigmented skin lesions due to autoimmune destruction of melanocytes. UVB phototherapy stimulates melanocyte stem cells in the hair bulge to undergo differentiation and upwards migration resulting in perifollicular repigmentation of vitiliginous lesions, which is under sophisticated signalling control. Melanocyte stem cells, normally quiescent, undergo cyclic activation/differentiation and downward migration with the hair cycle, providing pigment to hair follicles. Physiological hair greying results from progressive loss of melanocyte stem cells and can be accelerated by acute stress-induced, sympathetic driven hyperproliferation of the melanocyte stem cells. Ultimately, by reviewing the pathways governing epithelial and follicular pigmentation, numerous areas of future research and potential points of intervention are highlighted.


Assuntos
Folículo Piloso/fisiologia , Melanócitos/fisiologia , Fator de Transcrição Associado à Microftalmia/fisiologia , Pigmentação da Pele/fisiologia , Células-Tronco/fisiologia , Raios Ultravioleta , Vitiligo/terapia , Humanos , Terapia Ultravioleta/métodos
20.
Pigment Cell Melanoma Res ; 34(1): 28-43, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32777162

RESUMO

The second messenger cyclic adenosine monophosphate (cAMP) regulates numerous functions in both benign melanocytes and melanoma cells. cAMP is generated from two distinct sources, transmembrane and soluble adenylyl cyclases (tmAC and sAC, respectively), and is degraded by a family of proteins called phosphodiesterases (PDEs). cAMP signaling can be regulated in many different ways and can lead to varied effects in melanocytes. It was recently revealed that distinct cAMP signaling pathways regulate pigmentation by either altering pigment gene expression or the pH of melanosomes. In the context of melanoma, many studies report seemingly contradictory roles for cAMP in tumorigenesis. For example, cAMP signaling has been implicated in both cancer promotion and suppression, as well as both therapy resistance and sensitization. This conundrum in the field may be explained by the fact that cAMP signals in discrete microdomains and each microdomain can mediate differential cellular functions. Here, we review the role of cAMP signaling microdomains in benign melanocyte biology, focusing on pigmentation, and in melanomagenesis.


Assuntos
AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Melanócitos/citologia , Melanócitos/fisiologia , Melanoma/fisiopatologia , Transtornos da Pigmentação/fisiopatologia , Pigmentação , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...