Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Biochem Biophys Res Commun ; 684: 149066, 2023 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-37866241

RESUMO

Epidemiological studies have found that high citrus fruit consumption was associated with higher risk of skin cancer. Citrus fruits and some vegetables contain furocoumarins, which may interact with ultraviolet radiation to induce skin cancer. We aimed to determine the effects of two furocoumarins, including 8-methoxypsoralen (8-MOP) and 6',7'-dihydroxybergamottin (DHB), on UVA-induced DNA damage in human epidermal melanocytes, the origin of melanoma. Our hypothesis was that these dietary furocoumarins increase UVA-induced DNA damage in melanocytes, compared to cells exposed to UV alone. We incubated melanocytes with 8-MOP or DHB, followed by exposure to physiological doses of UVA radiation. We used Western blots to quantify the UVA-induced DNA damage measured by the fraction of phosphorylated histone variant H2AX (γH2AX), which is a marker of DNA damage, relative to total H2AX (γH2AX/H2AX) in the presence or absence of furocoumarins. To quantify the UVA-induced change in γH2AX/H2AX, we calculated the UVA:Control ratio as the ratio of γH2AX/H2AX in UVA-exposed cells to that in cells without UVA (control). The mean UVA:Control ratios were borderline significantly higher for cells treated with 8-MOP and significantly higher for cells treated with DHB, compared to that of untreated cells. This study suggests that furocoumarins (particularly 8-MOP and DHB) enhance UVA-induced DNA damage in melanocytes, which is a potential novel mechanism for citrus and furocoumarins to elevate the risk of skin cancer.


Assuntos
Citrus , Furocumarinas , Neoplasias Cutâneas , Humanos , Furocumarinas/farmacologia , Metoxaleno/farmacologia , Raios Ultravioleta/efeitos adversos , Melanócitos , Dano ao DNA
2.
Molecules ; 28(13)2023 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-37446583

RESUMO

Black poplar buds have high contents of many compounds with therapeutic potential, which are useful in cosmetics and the treatment of various dermatitis, respiratory diseases, etc. The aim of this study was to identify and exploit the local plant resources with biologically active properties from the Dobrogea area, Romania. For this purpose, materials were collected from the mentioned area, and macerates of black poplar were prepared in order to evaluate their qualities as antioxidant and antimicrobial agents. Three different black poplar buds' hydroalcoholic macerates were analyzed by the Folin-Ciocâlteau method to estimate the total content of phenolic compounds, by the HPLC-DAD method for identification and quantification of the main bioactive compounds and by the DPPH radical scavenging method to evaluate the antioxidant activity. All hydroalcoholic macerates showed high concentrations of phenolic compounds, the main individual compounds being gallic acid, chlorogenic acid, cinnamic acid, and methyl gallic acid. The antioxidant activity of the black poplar buds' hydroalcoholic macerates, evaluated by the DPPH radical scavenging test, showed high values, between 496 and 1200 mg GAE /100 g d.w. The Cd, Cu, Zn, Ni, and Pb concentrations released in dry poplar buds, determined by AAS, were below the detection limits. Hydroalcoholic macerates of black poplar were tested against two groups of gram-positive bacteria (Enterococcus and Staphylococcus) using an agar well diffusion assay. The in vitro inhibitory activities of the macerates were important and ranged from 8.2-9.4 mm inhibition zones (Staphylococcus) to 8.6 -10 mm inhibition zones (Enterococcus).


Assuntos
Anti-Infecciosos , Populus , Antioxidantes/farmacologia , Antioxidantes/química , Populus/química , Antibacterianos/farmacologia , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Anti-Infecciosos/farmacologia , Fenóis/farmacologia
3.
Proc Natl Acad Sci U S A ; 116(23): 11508-11517, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31097585

RESUMO

Opsins form a family of light-activated, retinal-dependent, G protein-coupled receptors (GPCRs) that serve a multitude of visual and nonvisual functions. Opsin 3 (OPN3 or encephalopsin), initially identified in the brain, remains one of the few members of the mammalian opsin family with unknown function and ambiguous light absorption properties. We recently discovered that OPN3 is highly expressed in human epidermal melanocytes (HEMs)-the skin cells that produce melanin. The melanin pigment is a critical defense against ultraviolet radiation (UVR), and its production is mediated by the Gαs-coupled melanocortin 1 receptor (MC1R). The physiological function and light sensitivity of OPN3 in melanocytes are yet to be determined. Here, we show that in HEMs, OPN3 acts as a negative regulator of melanin production by modulating the signaling of MC1R. OPN3 negatively regulates the cyclic adenosine monophosphate (cAMP) response evoked by MC1R via activation of the Gαi subunit of G proteins, thus decreasing cellular melanin levels. In addition to their functional relationship, OPN3 and MC1R colocalize at both the plasma membrane and in intracellular structures, and can form a physical complex. Remarkably, OPN3 can bind retinal, but does not mediate light-induced signaling in melanocytes. Our results identify a function for OPN3 in the regulation of the melanogenic pathway in epidermal melanocytes; we have revealed a light-independent function for the poorly characterized OPN3 and a pathway that greatly expands our understanding of melanocyte and skin physiology.


Assuntos
Epiderme/metabolismo , Melanócitos/metabolismo , Pigmentação/fisiologia , Receptor Tipo 1 de Melanocortina/metabolismo , Opsinas de Bastonetes/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Células HeLa , Humanos , Melaninas/metabolismo , Transdução de Sinais/fisiologia , Pele/metabolismo
4.
FASEB J ; 34(9): 11605-11623, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32658369

RESUMO

Exposure to high doses of solar long wavelength ultraviolet radiation (UVA) damages human skin via reactive oxygen species (ROS). Whether physiological UVA doses also generate ROS that has an effect on the skin remains unknown. We previously showed that in human epidermal melanocytes UVA activates a G-protein coupled signaling pathway that leads to calcium mobilization and increased melanin. Here, we report that ROS generated by the UVA phototransduction pathway are critical cellular messengers required to augment melanin. Using simultaneous UVA exposure and live-cell imaging of primary human melanocytes, we found that physiological doses of UVA generate two spatiotemporally distinct sources of ROS: one upstream of the G-protein activation that potentiates calcium responses, and another source downstream of calcium, in the mitochondria (ROSmito ). UVA-evoked signaling led to mitochondrial calcium uptake via mitochondrial calcium uniporter to promote ROSmito production leading to melanin synthesis. Our findings reveal a novel mechanism in which ROS function as signaling messengers necessary for melanin production, thus having a protective role in the UVA-induced skin response.


Assuntos
Cálcio/metabolismo , Melaninas/biossíntese , Melanócitos/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Raios Ultravioleta , Células Cultivadas , Prepúcio do Pênis/citologia , Humanos , Recém-Nascido , Masculino , Melanócitos/citologia , Melanócitos/metabolismo , Modelos Biológicos , Transdução de Sinais/efeitos da radiação
5.
Blood ; 132(19): 2053-2066, 2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30213875

RESUMO

Although the pathogenesis of primary myelofibrosis (PMF) and other myeloproliferative neoplasms (MPNs) is linked to constitutive activation of the JAK-STAT pathway, JAK inhibitors have neither curative nor MPN-stem cell-eradicating potential, indicating that other targetable mechanisms are contributing to the pathophysiology of MPNs. We previously demonstrated that Abelson interactor 1 (Abi-1), a negative regulator of Abelson kinase 1, functions as a tumor suppressor. Here we present data showing that bone marrow-specific deletion of Abi1 in a novel mouse model leads to development of an MPN-like phenotype resembling human PMF. Abi1 loss resulted in a significant increase in the activity of the Src family kinases (SFKs), STAT3, and NF-κB signaling. We also observed impairment of hematopoietic stem cell self-renewal and fitness, as evidenced in noncompetitive and competitive bone marrow transplant experiments. CD34+ hematopoietic progenitors and granulocytes from patients with PMF showed decreased levels of ABI1 transcript as well as increased activity of SFKs, STAT3, and NF-κB. In aggregate, our data link the loss of Abi-1 function to hyperactive SFKs/STAT3/NF-κB signaling and suggest that this signaling axis may represent a regulatory module involved in the molecular pathophysiology of PMF.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Medula Óssea/patologia , Proteínas do Citoesqueleto/genética , Deleção de Genes , Mielofibrose Primária/genética , Mielofibrose Primária/patologia , Animais , Medula Óssea/metabolismo , Autorrenovação Celular , Células Cultivadas , Regulação para Baixo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Mielofibrose Primária/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(6): 2383-8, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23345429

RESUMO

Human skin is constantly exposed to solar ultraviolet radiation (UVR), the most prevalent environmental carcinogen. Humans have the unique ability among mammals to respond to UVR by increasing their skin pigmentation, a protective process driven by melanin synthesis in epidermal melanocytes. The molecular mechanisms used by melanocytes to detect and respond to long-wavelength UVR (UVA) are not well understood. We recently identified a UVA phototransduction pathway in melanocytes that is mediated by G protein-coupled receptors and leads to rapid calcium mobilization. Here we report that in human epidermal melanocytes physiological doses of UVR activate a retinal-dependent current mediated by transient receptor potential A1 (TRPA1) ion channels. The TRPA1 photocurrent is UVA-specific and requires G protein and phospholipase C signaling, thus contributing to UVA-induced calcium responses to mediate downstream cellular effects and providing evidence for TRPA1 function in mammalian phototransduction. Remarkably, TRPA1 activation is required for the UVR-induced and retinal-dependent early increase in cellular melanin. Our results show that TRPA1 is essential for a unique extraocular phototransduction pathway in human melanocytes that is activated by physiological doses of UVR and results in early melanin synthesis.


Assuntos
Canais de Cálcio/metabolismo , Canais de Cálcio/efeitos da radiação , Transdução de Sinal Luminoso/fisiologia , Transdução de Sinal Luminoso/efeitos da radiação , Melanócitos/metabolismo , Melanócitos/efeitos da radiação , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/efeitos da radiação , Canais de Potencial de Receptor Transitório/metabolismo , Canais de Potencial de Receptor Transitório/efeitos da radiação , Animais , Células CHO , Canais de Cálcio/genética , Sinalização do Cálcio/efeitos da radiação , Células Cultivadas , Cricetinae , Cricetulus , Proteínas de Ligação ao GTP/metabolismo , Humanos , Melaninas/biossíntese , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/genética , Raios Ultravioleta
7.
Genomics ; 104(6 Pt B): 482-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25451175

RESUMO

Because human epidermal melanocytes (HEMs) provide critical protection against skin cancer, sunburn, and photoaging, a genome-wide perspective of gene expression in these cells is vital to understanding human skin physiology. In this study we performed high throughput sequencing of HEMs to obtain a complete data set of transcript sizes, abundances, and splicing. As expected, we found that melanocyte specific genes that function in pigmentation were among the highest expressed genes. We analyzed receptor, ion channel and transcription factor gene families to get a better understanding of the cell signaling pathways used by melanocytes. We also performed a comparative transcriptomic analysis of lightly versus darkly pigmented HEMs and found 16 genes differentially expressed in the two pigmentation phenotypes; of those, only one putative melanosomal transporter (SLC45A2) has known function in pigmentation. In addition, we found 166 transcript isoforms expressed exclusively in one pigmentation phenotype, 17 of which are genes involved in signal transduction. Our melanocyte transcriptome study provides a comprehensive view and may help identify novel pigmentation genes and potential pharmacological targets.


Assuntos
Epiderme/metabolismo , Genoma Humano , Melanócitos/metabolismo , Pigmentação da Pele/genética , Transcriptoma , Linhagem Celular , Células Epidérmicas , Perfilação da Expressão Gênica , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Arch Biochem Biophys ; 563: 35-41, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25034214

RESUMO

Skin melanocytes and ocular pigment cells contain specialized organelles called melanosomes, which are responsible for the synthesis of melanin, the major pigment in mammals. Defects in the complex mechanisms involved in melanin synthesis and regulation result in vision and pigmentation deficits, impaired development of the visual system, and increased susceptibility to skin and eye cancers. Ion transport across cellular membranes is critical for many biological processes, including pigmentation, but the molecular mechanisms by which it regulates melanin synthesis, storage, and transfer are not understood. In this review we first discuss ion channels and transporters that function at the plasma membrane of melanocytes; in the second part we consider ion transport across the membrane of intracellular organelles, with emphasis on melanosomes. We discuss recently characterized lysosomal and endosomal ion channels and transporters associated with pigmentation phenotypes. We then review the evidence for melanosomal channels and transporters critical for pigmentation, discussing potential molecular mechanisms mediating their function. The studies investigating ion transport in pigmentation physiology open new avenues for future research and could reveal novel molecular mechanisms underlying melanogenesis.


Assuntos
Transporte de Íons/fisiologia , Pigmentação/fisiologia , Animais , Sinalização do Cálcio , Endossomos/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Canais Iônicos/metabolismo , Lisossomos/metabolismo , Melaninas/biossíntese , Melanócitos/metabolismo , Melanossomas/metabolismo , Potenciais da Membrana
9.
Nat Genet ; 56(2): 258-272, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38200130

RESUMO

Skin color is highly variable in Africans, yet little is known about the underlying molecular mechanism. Here we applied massively parallel reporter assays to screen 1,157 candidate variants influencing skin pigmentation in Africans and identified 165 single-nucleotide polymorphisms showing differential regulatory activities between alleles. We combine Hi-C, genome editing and melanin assays to identify regulatory elements for MFSD12, HMG20B, OCA2, MITF, LEF1, TRPS1, BLOC1S6 and CYB561A3 that impact melanin levels in vitro and modulate human skin color. We found that independent mutations in an OCA2 enhancer contribute to the evolution of human skin color diversity and detect signals of local adaptation at enhancers of MITF, LEF1 and TRPS1, which may contribute to the light skin color of Khoesan-speaking populations from Southern Africa. Additionally, we identified CYB561A3 as a novel pigmentation regulator that impacts genes involved in oxidative phosphorylation and melanogenesis. These results provide insights into the mechanisms underlying human skin color diversity and adaptive evolution.


Assuntos
Albinismo Oculocutâneo , Melaninas , Pigmentação da Pele , Humanos , Pigmentação da Pele/genética , Melaninas/genética , Alelos , Genômica , Pigmentação/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Repressoras/genética
10.
Adv Exp Med Biol ; 704: 135-45, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21290293

RESUMO

TRPM1, initially named Melastatin, is the founding member of the TRPM subfamily of Transient Receptor Potential (TRP) ion channels. Despite sustained efforts, the molecular properties and physiological functions of TRPM1 remained elusive until recently. New evidence has uncovered novel TRPM1 splice variants and revealed that TRPM1 is critical for a non-selective cation conductance in melanocytes and retinal bipolar cells. Functionally, TRPM1 has been shown to mediate retinal ON bipolar cell transduction and suggested to regulate melanocyte pigmentation. Notably, TRPM1 mutations have also been associated with congenital stationary night blindness in humans. This review will summarize and discuss our present knowledge of TRPM1: its discovery, expression, regulation, and proposed functions in skin and eye.


Assuntos
Canais de Cátion TRPM/fisiologia , Regulação da Expressão Gênica , Humanos , Mutação , Cegueira Noturna/congênito , Cegueira Noturna/genética , Splicing de RNA , RNA Mensageiro/genética , Frações Subcelulares/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
11.
Methods Enzymol ; 654: 315-344, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34120720

RESUMO

Melanocytes are specialized cells that produce melanin pigments responsible for skin, hair, and eye pigmentation. The synthesis and storage of melanin occurs in unique lysosome-related organelles called melanosomes, which regulate melanin production via complex regulatory mechanisms. Maintenance of the melanosome luminal ionic environment and pH is crucial for proper function of the main melanogenic enzymes. Defects in genes encoding pH-regulating melanosomal proteins result in oculocutaneous albinism, which is characterized by hypopigmentation, impaired vision, and increased susceptibility to skin and eye cancers. We recently uncovered several ion channels and transporters that modulate melanin synthesis by acidifying or neutralizing the luminal pH of melanosomes. However, our understanding of how melanosomes and other related organelles maintain their luminal pH is far from complete. The study of melanosome pH regulation requires robust imaging and quantification tools. Despite recent advances in the development of such methods, many limitations remain, particularly for quantitative analysis of individual organelle pH. In this chapter, we will provide an overview of the available methods used for melanosome pH determination, including their advantages, limitations, and challenges. To address the critical, unmet need for reliable melanosome pH quantification tools, we engineered a novel genetically encoded, ratiometric pH sensor for melanosomes that we named RpHiMEL. Here, we describe the design and optimization of RpHiMEL, and provide a pH quantification method for individual melanosomes in live cells. We demonstrate that RpHiMEL is a highly versatile tool with the potential to advance our understanding of pH regulation in melanosomes and related organelles.


Assuntos
Melanócitos , Melanossomas , Concentração de Íons de Hidrogênio , Melaninas , Pigmentação
12.
Mol Cell Biol ; 27(4): 1486-94, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17178838

RESUMO

Extracellular pH has long been known to affect the rate and magnitude of ion transport processes among others via regulation of ion channel activity. The Ca(2+)-selective transient receptor potential vanilloid 5 (TRPV5) channel constitutes the apical entry gate in Ca(2+)-transporting cells, contributing significantly to the overall Ca(2+) balance. Here, we demonstrate that extracellular pH determines the cell surface expression of TRPV5 via a unique mechanism. By a comprehensive approach using total internal reflection fluorescence microscopy, cell surface protein labeling, electrophysiology, (45)Ca(2+) uptake assays, and functional channel recovery after chemobleaching, this study shows that upon extracellular alkalinization, a pool of TRPV5-containing vesicles is rapidly recruited to the cell surface without collapsing into the plasma membrane. These vesicles contain functional TRPV5 channels since extracellular alkalinization is accompanied by increased TRPV5 activity. Conversely, upon subsequent extracellular acidification, vesicles are retrieved from the plasma membrane, simultaneously resulting in decreased TRPV5 activity. Thus, TRPV5 accesses the extracellular compartment via transient openings of vesicles, suggesting that rapid responses of constitutive active TRP channels to physiological stimuli rely on vesicular "kiss and linger" interactions with the plasma membrane.


Assuntos
Membrana Celular/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Cálcio/metabolismo , Cães , Humanos , Concentração de Íons de Hidrogênio , Transporte Proteico , Vesículas Transportadoras/metabolismo
13.
Adv Biol Regul ; 75: 100668, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31653550

RESUMO

Because sunlight is essential for human survival, we have developed complex mechanisms for detecting and responding to light stimuli. The eyes and skin are major organs for sensing light and express several light-sensitive opsin receptors. These opsins mediate cellular responses to spectrally-distinct wavelengths of visible and ultraviolet light. How the eyes mediate visual phototransduction is well understood, but less is known about how the skin detects light. Both human and murine skin express a wide array of opsins, with one of the most highly expressed being the functionally elusive opsin 3 (OPN3). In this review we explore light reception, opsin expression and signaling in skin cells; we compile data elucidating potential functions for human OPN3 in skin, with emphasis on recent studies investigating OPN3 regulation of melanin within epidermal melanocytes.


Assuntos
Epiderme/metabolismo , Regulação da Expressão Gênica/fisiologia , Melanócitos/metabolismo , Opsinas de Bastonetes/biossíntese , Transdução de Sinais/fisiologia , Animais , Humanos , Camundongos
14.
eNeuro ; 7(5)2020.
Artigo em Inglês | MEDLINE | ID: mdl-32737180

RESUMO

The opsins have been studied extensively for their functions in visual phototransduction; however, the mechanisms underlying extraocular opsin signaling remain poorly understood. The first mammalian extraocular opsin to be discovered, opsin 3 (OPN3), was found in the brain more than two decades ago, yet its function remains unknown. A significant hindrance to studying OPN3 has been a lack of specific antibodies against mammalian OPN3, resulting in an incomplete understanding of its expression in the brain. Although Opn3 promoter-driven reporter mice have been generated to examine general OPN3 localization, they lack the regulated expression of the endogenous protein and the ability to study its subcellular localization. To circumvent these issues, we have created a knock-in OPN3-mCherry mouse model in which the fusion protein OPN3-mCherry is expressed under the endogenous Opn3 promoter. Viable and fertile homozygotes for the OPN3-mCherry allele were used to create an extensive map of OPN3-mCherry expression across the adult mouse brain. OPN3-mCherry was readily visualized in distinct layers of the cerebral cortex (CTX), the hippocampal formation (HCF), distinct nuclei of the thalamus, as well as many other regions in both neuronal and non-neuronal cells. Our mouse model offers a new platform to investigate the function of OPN3 in the brain.


Assuntos
Opsinas , Opsinas de Bastonetes , Animais , Encéfalo/metabolismo , Camundongos , Opsinas/genética , Opsinas de Bastonetes/metabolismo , Transdução de Sinais
15.
Mol Biol Cell ; 31(24): 2687-2702, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32966160

RESUMO

SLC45A2 encodes a putative transporter expressed primarily in pigment cells. SLC45A2 mutations cause oculocutaneous albinism type 4 (OCA4) and polymorphisms are associated with pigmentation variation, but the localization, function, and regulation of SLC45A2 and its variants remain unknown. We show that SLC45A2 localizes to a cohort of mature melanosomes that only partially overlaps with the cohort expressing the chloride channel OCA2. SLC45A2 expressed ectopically in HeLa cells localizes to lysosomes and raises lysosomal pH, suggesting that in melanocytes SLC45A2 expression, like OCA2 expression, results in the deacidification of maturing melanosomes to support melanin synthesis. Interestingly, OCA2 overexpression compensates for loss of SLC45A2 expression in pigmentation. Analyses of SLC45A2- and OCA2-deficient mouse melanocytes show that SLC45A2 likely functions later during melanosome maturation than OCA2. Moreover, the light skin-associated SLC45A2 allelic F374 variant restores only moderate pigmentation to SLC45A2-deficient melanocytes due to rapid proteasome-dependent degradation resulting in lower protein expression levels in melanosomes than the dark skin-associated allelic L374 variant. Our data suggest that SLC45A2 maintains melanosome neutralization that is initially orchestrated by transient OCA2 activity to support melanization at late stages of melanosome maturation, and that a common allelic variant imparts reduced activity due to protein instability.


Assuntos
Antígenos de Neoplasias/metabolismo , Melanócitos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Pigmentação da Pele/fisiologia , Animais , Antígenos de Neoplasias/fisiologia , Proteínas de Transporte/metabolismo , Linhagem Celular , Canais de Cloreto/metabolismo , Células HeLa , Humanos , Lisossomos/metabolismo , Masculino , Melanossomas/metabolismo , Proteínas de Membrana Transportadoras/fisiologia , Camundongos , Pigmentação/fisiologia , Estabilidade Proteica , Pele/metabolismo
16.
Dev Cell ; 4(4): 561-74, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12689594

RESUMO

The specificity of many signal transduction pathways relies on the spatiotemporal features of each signaling step. G protein-coupled receptor-mediated activation of protein kinases leads to diverse cellular effects. Upon receptor activation, PKD1 and several C-type protein kinases (PKCs), translocate to the plasma membrane and become catalytically active. Here we show that, unlike PKCs, PKD1 remains active at the membrane for hours. The two DAG binding C1 domains of PKD1 have distinct functional roles in targeting and maintaining PKD1 at the plasma membrane. C1A achieves fast, maximal, and reversible translocation, while C1B translocates partially, but persistently, to the plasma membrane. The persistent localization requires the C1B domain of PKD1, which binds Galphaq. We incorporate the kinetics of PKD1 translocation into a three-state model that suggests how PKD1 binding to DAG and Galphaq uniquely encodes frequency-dependent PKD1 signaling.


Assuntos
Membrana Celular/enzimologia , Células Eucarióticas/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Proteína Quinase C/metabolismo , Transporte Proteico/fisiologia , Receptores de Superfície Celular/metabolismo , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Diglicerídeos/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Hormônios/metabolismo , Hormônios/farmacologia , Humanos , Modelos Biológicos , Mutação/genética , Ligação Proteica/fisiologia , Isoformas de Proteínas/metabolismo , Proteína Quinase C/genética , Estrutura Terciária de Proteína/fisiologia , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Proteínas Recombinantes de Fusão , Transdução de Sinais/fisiologia
17.
Circ Res ; 98(2): 245-53, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16357306

RESUMO

Many cells are constantly exposed to fluid mechanical forces generated by flowing blood, and wall shear stresses modulate aspects of their structure and function. However, the mechanisms for mechanotransduction of flow are not well understood. Here we report that TRPM7, which is both an ion channel and a functional kinase, is translocated within cells in response to laminar flow. After exposure of cells to physiological values of laminar fluid flow, the number of TRPM7 molecules localized at or near the plasma membrane increased up to 2-fold, in less than 100 seconds. This increase in membrane-localized GFP-TRPM7, as seen by total internal reflection fluorescence microscopy, closely correlated with increases in TRPM7 current. Both endogenous and heterologously expressed TRPM7 was found in tubulovesicular structures that were translocated to the region of the plasma membrane on induction of shear stress. In vascular smooth muscle cells, but not in several types of endothelial cells, fluid flow increased endogenous native TRPM7 current amplitude. We hypothesize that TRPM7 plays a role in pathological response to vessel wall injury.


Assuntos
Circulação Sanguínea , Membrana Celular/metabolismo , Músculo Liso Vascular/metabolismo , Canais de Cátion TRPM/fisiologia , Células Cultivadas , Humanos , Músculo Liso Vascular/química , Músculo Liso Vascular/citologia , Proteínas Serina-Treonina Quinases , Transporte Proteico , Resistência ao Cisalhamento , Estresse Mecânico , Canais de Cátion TRPM/análise
18.
Nat Neurosci ; 6(8): 837-45, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12858178

RESUMO

Growth cone motility is regulated by both fast voltage-dependent Ca2+ channels and by unknown receptor-operated Ca2+ entry mechanisms. Transient receptor potential (TRP) homomeric TRPC5 ion channels are receptor-operated, Ca2+-permeable channels predominantly expressed in the brain. Here we show that TRPC5 is expressed in growth cones of young rat hippocampal neurons. Our results indicate that TRPC5 channel subunits interact with the growth cone-enriched protein stathmin 2, are packaged into vesicles and are carried to newly forming growth cones and synapses. Once in the growth cone, TRPC5 channels regulate neurite extension and growth-cone morphology. Dominant-negative TRPC5 expression allowed significantly longer neurites and filopodia to form. We conclude that TRPC5 channels are important components of the mechanism controlling neurite extension and growth cone morphology.


Assuntos
Canais de Cálcio/fisiologia , Proteínas de Transporte de Cátions , Cones de Crescimento/ultraestrutura , Hipocampo/ultraestrutura , Proteínas dos Microtúbulos , Neuritos/ultraestrutura , Animais , Canais de Cálcio/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Condutividade Elétrica , Hipocampo/metabolismo , Imuno-Histoquímica , Microscopia Eletrônica , Neurônios/metabolismo , Fosfoproteínas/fisiologia , Isoformas de Proteínas/fisiologia , Ratos , Estatmina , Canais de Cátion TRPC
19.
Sci Rep ; 6: 26570, 2016 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-27231233

RESUMO

Intracellular organelles mediate complex cellular functions that often require ion transport across their membranes. Melanosomes are organelles responsible for the synthesis of the major mammalian pigment melanin. Defects in melanin synthesis result in pigmentation defects, visual deficits, and increased susceptibility to skin and eye cancers. Although genes encoding putative melanosomal ion transporters have been identified as key regulators of melanin synthesis, melanosome ion transport and its contribution to pigmentation remain poorly understood. Here we identify two-pore channel 2 (TPC2) as the first reported melanosomal cation conductance by directly patch-clamping skin and eye melanosomes. TPC2 has been implicated in human pigmentation and melanoma, but the molecular mechanism mediating this function was entirely unknown. We demonstrate that the vesicular signaling lipid phosphatidylinositol bisphosphate PI(3,5)P2 modulates TPC2 activity to control melanosomal membrane potential, pH, and regulate pigmentation.


Assuntos
Canais de Cálcio/metabolismo , Olho/citologia , Melanossomas/metabolismo , Pigmentação , Pele/citologia , Animais , Canais de Cálcio/genética , Células Cultivadas , Olho/metabolismo , Concentração de Íons de Hidrogênio , Melanócitos/citologia , Melanócitos/metabolismo , Melanossomas/fisiologia , Potenciais da Membrana , Camundongos , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Pele/metabolismo
20.
Sci Signal ; 9(444): ra87, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27601729

RESUMO

Bone morphogenetic proteins (BMPs) function in most tissues but have cell type-specific effects. Given the relatively small number of BMP receptors, this exquisite signaling specificity requires additional molecules to regulate this pathway's output. The receptor tyrosine kinase MuSK (muscle-specific kinase) is critical for neuromuscular junction formation and maintenance. Here, we show that MuSK also promotes BMP signaling in muscle cells. MuSK bound to BMP4 and related BMPs with low nanomolar affinity in vitro and to the type I BMP receptors ALK3 and ALK6 in a ligand-independent manner both in vitro and in cultured myotubes. High-affinity binding to BMPs required the third, alternatively spliced MuSK immunoglobulin-like domain. In myoblasts, endogenous MuSK promoted BMP4-dependent phosphorylation of SMADs and transcription of Id1, which encodes a transcription factor involved in muscle differentiation. Gene expression profiling showed that MuSK was required for the BMP4-induced expression of a subset of genes in myoblasts, including regulator of G protein signaling 4 (Rgs4). In myotubes, MuSK enhanced the BMP4-induced expression of a distinct set of genes, including transcripts characteristic of slow muscle. MuSK-mediated stimulation of BMP signaling required type I BMP receptor activity but was independent of MuSK tyrosine kinase activity. MuSK-dependent expression of Rgs4 resulted in the inhibition of Ca(2+) signaling induced by the muscarinic acetylcholine receptor in myoblasts. These findings establish that MuSK has dual roles in muscle cells, acting both as a tyrosine kinase-dependent synaptic organizing molecule and as a BMP co-receptor that shapes BMP transcriptional output and cholinergic signaling.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Mioblastos/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Colinérgicos/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteína Morfogenética Óssea 4/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Linhagem Celular , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Camundongos , Mioblastos/citologia , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Colinérgicos/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA