Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cancer Res ; 81(23): 5977-5990, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34642183

RESUMO

The relationship between cancer and autoimmunity is complex. However, the incidence of solid tumors such as melanoma has increased significantly among patients with previous or newly diagnosed systemic autoimmune disease (AID). At the same time, immune checkpoint blockade (ICB) therapy of cancer induces de novo autoinflammation and exacerbates underlying AID, even without evident antitumor responses. Recently, systemic lupus erythematosus (SLE) activity was found to drive myeloid-derived suppressor cell (MDSC) formation in patients, a known barrier to healthy immune surveillance and successful cancer immunotherapy. Cross-talk between MDSCs and macrophages generally drives immune suppressive activity in the tumor microenvironment. However, it remains unclear how peripheral pregenerated MDSC under chronic inflammatory conditions modulates global macrophage immune functions and the impact it could have on existing tumors and underlying lupus nephritis. Here we show that pathogenic expansion of SLE-generated MDSCs by melanoma drives global macrophage polarization and simultaneously impacts the severity of lupus nephritis and tumor progression in SLE-prone mice. Molecular and functional data showed that MDSCs interact with autoimmune macrophages and inhibit cell surface expression of CD40 and the production of IL27. Moreover, low CD40/IL27 signaling in tumors correlated with high tumor-associated macrophage infiltration and ICB therapy resistance both in murine and human melanoma exhibiting active IFNγ signatures. These results suggest that preventing global macrophage reprogramming induced by MDSC-mediated inhibition of CD40/IL27 signaling provides a precision melanoma immunotherapy strategy, supporting an original and advantageous approach to treat solid tumors within established autoimmune landscapes. SIGNIFICANCE: Myeloid-derived suppressor cells induce macrophage reprogramming by suppressing CD40/IL27 signaling to drive melanoma progression, simultaneously affecting underlying autoimmune disease and facilitating resistance to immunotherapy within preexisting autoimmune landscapes.


Assuntos
Autoimunidade , Antígenos CD40/metabolismo , Interleucina-27/metabolismo , Lúpus Eritematoso Sistêmico/fisiopatologia , Macrófagos/patologia , Melanoma/patologia , Células Supressoras Mieloides/patologia , Animais , Imunoterapia , Macrófagos/imunologia , Macrófagos/metabolismo , Melanoma/imunologia , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Microambiente Tumoral
2.
J Interferon Cytokine Res ; 39(1): 72-84, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30562133

RESUMO

Today, improvements in diagnostic and therapeutic options allow patients with autoimmune diseases (ADs) to live longer and have more active lives compared with patients receiving conventional anti-inflammatory therapy just two decades ago. Current therapies for ADs aim to inhibit immune cell activation and effector immune pathways, including those activated by cytokines and cytokine receptors. Understandably, such goals become more complicated in patients with long-term established ADs who develop parallel chronic or comorbid conditions, including life-threatening diseases, such as cancer. Compared with the general population, patients with ADs have an increased risk of developing hematological, lymphoproliferative disorders, and solid tumors. However, the aim of current cancer therapies is to activate the immune system to create autoimmune-like conditions and eliminate tumors. As such, their comorbid presentation creates a paradox on how malignancies must be addressed therapeutically in the context of autoimmunity. Because the physiopathology of malignancies is less understood in the context of autoimmunity than it is in the general population, we undertook this review to highlight the peculiarities and mechanisms governing immune cells in established ADs. Moreover, we examined the role of the autoimmune cytokine milieu in the development of immune-related adverse events during the implementation of conventional or immune-based therapy.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Citocinas/imunologia , Neoplasias/imunologia , Doenças Autoimunes/terapia , Humanos , Imunoterapia , Neoplasias/terapia
3.
J Biol Chem ; 292(34): 13925-13933, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28652404

RESUMO

Interferon γ (IFNγ) is a pleiotropic protein secreted by immune cells. IFNγ signals through the IFNγ receptor, a protein complex that mediates downstream signaling events. Studies into IFNγ signaling have provided insight into the general concepts of receptor signaling, receptor internalization, regulation of distinct signaling pathways, and transcriptional regulation. Although IFNγ is the central mediator of the adaptive immune response to pathogens, it has been shown to be involved in several non-infectious physiological processes. This review will provide an introduction into IFNγ signaling biology and the functional roles of IFNγ in the autoimmune response.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Autoimunidade , Interferon gama/metabolismo , Modelos Biológicos , Receptores de Interferon/agonistas , Transdução de Sinais , Animais , Células Apresentadoras de Antígenos/imunologia , Autofagossomos/imunologia , Autofagossomos/metabolismo , Cavéolas/imunologia , Cavéolas/metabolismo , Vesículas Revestidas por Clatrina/imunologia , Vesículas Revestidas por Clatrina/metabolismo , Dimerização , Endocitose , Humanos , Interferon gama/química , Macrófagos/imunologia , Macrófagos/metabolismo , Microdomínios da Membrana , Multimerização Proteica , Receptores de Interferon/química , Receptores de Interferon/metabolismo , Receptor de Interferon gama
4.
Cancer Res ; 75(13): 2708-15, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25832654

RESUMO

The AMPK-related kinase NUAK2 has been implicated in melanoma growth and survival outcomes, but its therapeutic utility has yet to be confirmed. In this study, we show how its genetic amplification in PTEN-deficient melanomas may rationalize the use of CDK2 inhibitors as a therapeutic strategy. Analysis of array-CGH data revealed that PTEN deficiency is coupled tightly with genomic amplification encompassing the NUAK2 locus, a finding strengthened by immunohistochemical evidence that phospho-Akt overexpression was correlated with NUAK2 expression in clinical specimens of acral melanoma. Functional studies in melanoma cells showed that inactivation of the PI3K pathway upregulated p21 expression and reduced the number of cells in S phase. NUAK2 silencing and inactivation of the PI3K pathway efficiently controlled CDK2 expression, whereas CDK2 inactivation specifically abrogated the growth of NUAK2-amplified and PTEN-deficient melanoma cells. Immunohistochemical analyses confirmed an association of CDK2 expression with NUAK2 amplification and p-Akt expression in melanomas. Finally, pharmacologic inhibition of CDK2 was sufficient to suppress the growth of NUAK2-amplified and PTEN-deficient melanoma cells in vitro and in vivo. Overall, our results show how CDK2 blockade may offer a promising therapy for genetically defined melanomas, where NUAK2 is amplified and PTEN is deleted.


Assuntos
Quinase 2 Dependente de Ciclina/metabolismo , Melanoma/genética , PTEN Fosfo-Hidrolase/deficiência , Proteínas Serina-Treonina Quinases/genética , Neoplasias Cutâneas/genética , Idoso , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Amplificação de Genes , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Melanoma/patologia , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Terapia de Alvo Molecular , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Roscovitina , Transdução de Sinais , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia
5.
Chest ; 147(3): 771-777, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25411763

RESUMO

BACKGROUND: Lymphangioleiomyomatosis (LAM) is characterized by the proliferation in the lung, axial lymphatics (eg, lymphangioleiomyomas), and kidney (eg, angiomyolipomas) of abnormal smooth muscle-like LAM cells, which express melanoma antigens such as Pmel17/gp100 and have dysfunctional tumor suppressor tuberous sclerosis complex (TSC) genes TSC2 or TSC1. Histopathologic diagnosis of LAM in lung specimens is based on identification of the Pmel17 protein with the monoclonal antibody HMB-45. METHODS: We compared the sensitivity of HMB-45 to that of antipeptide antibody αPEP13h, which reacts with a C-terminal peptide of Pmel17. LAM lung nodules were laser-capture microdissected to identify proteins by Western blotting. RESULTS: HMB-45 recognized approximately 25% of LAM cells within the LAM lung nodules, whereas αPEP13h identified > 82% of LAM cells within these structures in approximately 90% of patients. Whereas HMB-45 reacted with epithelioid but not with spindle-shaped LAM cells, αPEP13h identified both spindle-shaped and epithelioid LAM cells, providing greater sensitivity for detection of all types of LAM cells. HMB-45 recognized Pmel17 in premelanosomal organelles; αPEP13h recognized proteins in the cytoplasm as well as in premelanosomal organelles. Both antibodies recognized a Pmel17 variant of approximately 50 kDa. CONCLUSIONS: Based on its sensitivity and specificity, αPEP13h may be useful in the diagnosis of LAM and more sensitive than HMB-45.


Assuntos
Anticorpos Anti-Idiotípicos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/patologia , Linfangioleiomiomatose/diagnóstico , Linfangioleiomiomatose/patologia , Nódulo Pulmonar Solitário/diagnóstico , Nódulo Pulmonar Solitário/patologia , Adulto , Anticorpos Anti-Idiotípicos/imunologia , Biópsia , Broncoscopia , Células Cultivadas , Diagnóstico Diferencial , Feminino , Humanos , Imuno-Histoquímica , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Linfangioleiomiomatose/imunologia , Antígenos Específicos de Melanoma/imunologia , Pessoa de Meia-Idade , Sensibilidade e Especificidade , Nódulo Pulmonar Solitário/imunologia , Antígeno gp100 de Melanoma/imunologia
6.
J Dermatol Sci ; 77(1): 21-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25530116

RESUMO

BACKGROUND: Patients with oculocutaneous albinism (OCA) have severely decreased pigmentation of their skin, hair and eyes. OCA2 and OCA4 result from mutations of the OCA2 and SLC45A2 genes, respectively, both of which disrupt the trafficking of the critical melanogenic enzyme tyrosinase to melanosomes. Both proteins encoded by those loci (termed P and MATP, respectively) have 12 putative transmembrane regions and are thought to function as transporters, although their functions and subcellular localizations remain to be characterized. OBJECTIVE: To generate specific antibodies against unique synthetic peptides encoded by P and MATP that could be used to characterize their functions and subcellular localizations. METHODS: Western blotting and immunohistochemistry were used to assess the specificity of antibodies and to colocalize P and MATP proteins with various subcellular markers. RESULTS: Specific antibodies to the P and MATP proteins were generated that work well for Western blotting and immunohistochemistry. The localizations of P and MATP with various subcellular organelles were characterized using confocal microscopy, which revealed that they colocalize to some extent with LAMP2, but do not significantly colocalize with markers of the ER, Golgi or melanosomes. Interestingly, both P and MATP colocalize significantly with BLOC-1, a sorting component involved in the intracellular trafficking of melanosomal/lysosomal constituents. CONCLUSION: These results provide a basis to understand how disrupted functions of P or MATP result in the misrouting of tyrosinase and cause the hypopigmentation seen in OCA2 and OCA4.


Assuntos
Albinismo Oculocutâneo/imunologia , Anticorpos/química , Hipopigmentação/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Sequência de Aminoácidos , Antígenos de Neoplasias/metabolismo , Transporte Biológico , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Humanos , Imuno-Histoquímica , Masculino , Melanócitos/citologia , Melanossomas/imunologia , Melanossomas/metabolismo , Dados de Sequência Molecular , Monofenol Mono-Oxigenase/imunologia , Monofenol Mono-Oxigenase/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/química , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo
7.
J Pathol ; 236(1): 17-29, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25488118

RESUMO

Human skin colour, ie pigmentation, differs widely among individuals, as do their responses to various types of ultraviolet radiation (UV) and their risks of skin cancer. In some individuals, UV-induced pigmentation persists for months to years in a phenomenon termed long-lasting pigmentation (LLP). It is unclear whether LLP is an indicator of potential risk for skin cancer. LLP seems to have similar features to other forms of hyperpigmentation, eg solar lentigines or age spots, which are clinical markers of photodamage and risk factors for precancerous lesions. To investigate what UV-induced molecular changes may persist in individuals with LLP, clinical specimens from non-sunburn-inducing repeated UV exposures (UVA, UVB or UVA + UVB) at 4 months post-exposure (short-term LLP) were evaluated by microarray analysis and dataset mining. Validated targets were further evaluated in clinical specimens from six healthy individuals (three LLP+ and three LLP-) followed for more than 9 months (long-term LLP) who initially received a single sunburn-inducing UVA + UVB exposure. The results support a UV-induced hyperpigmentation model in which basal keratinocytes have an impaired ability to remove melanin that leads to a compensatory mechanism by neighbouring keratinocytes with increased proliferative capacity to maintain skin homeostasis. The attenuated expression of SOX7 and other hemidesmosomal components (integrin α6ß4 and plectin) leads to increased melanosome uptake by keratinocytes and points to a spatial regulation within the epidermis. The reduced density of hemidesmosomes provides supporting evidence for plasticity at the epidermal-dermal junction. Altered hemidesmosome plasticity, and the sustained nature of LLP, may be mediated by the role of SOX7 in basal keratinocytes. The long-term sustained subtle changes detected are modest, but sufficient to create dramatic visual differences in skin colour. These results suggest that the hyperpigmentation phenomenon leading to increased interdigitation develops in order to maintain normal skin homeostasis in individuals with LLP.


Assuntos
Epiderme/metabolismo , Hemidesmossomos/metabolismo , Queratinócitos/metabolismo , Pigmentação da Pele/efeitos da radiação , Pele/metabolismo , Raios Ultravioleta/efeitos adversos , Células Cultivadas , Epiderme/efeitos da radiação , Hemidesmossomos/efeitos da radiação , Humanos , Queratinócitos/efeitos da radiação , Pele/efeitos da radiação , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Tempo
8.
Pigment Cell Melanoma Res ; 27(1): 82-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24024552

RESUMO

Through a process known as melanogenesis, melanocyte produces melanin in specialized organelles termed melanosomes, which regulates pigmentation of the skin, eyes, and hair. Gp96 is a constitutively expressed heat shock protein in the endoplasmic reticulum whose expression is further upregulated upon ultraviolet irradiation. However, the roles and mechanisms of this chaperone in pigmentation biology are unknown. In this study, we found that knockdown of gp96 by RNA interference significantly perturbed melanin synthesis and blocked late melanosome maturation. Gp96 knockdown did not impair the expression of tyrosinase, an essential enzyme in melanin synthesis, but compromised its catalytic activity and melanosome translocation. Further, mice with melanocyte-specific deletion of gp96 displayed decreased pigmentation. A mechanistic study revealed that the defect in melanogenesis can be rescued by activation of the canonical Wnt pathway, consistent with the critical roles of gp96 in chaperoning Wnt-coreceptor LRP6. Thus, this work uncovered the essential role of gp96 in regulating melanogenesis.


Assuntos
Regulação da Expressão Gênica/fisiologia , Melaninas/biossíntese , Melanossomas/metabolismo , Glicoproteínas de Membrana/biossíntese , Pigmentação da Pele/fisiologia , Animais , Linhagem Celular , Melaninas/genética , Melanossomas/genética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos
9.
J Invest Dermatol ; 132(12): 2791-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22895365

RESUMO

Diacylglycerol (DAG) increases the melanin content of human melanocytes in vitro and increases the pigmentation of guinea pig skin in vivo, but the mechanism(s) underlying those effects remain unknown. In this study, we characterized the role of diacylglycerol kinase (DGK), which phosphorylates DAG to generate phosphatidic acid, in the regulation of pigmentation. Ten isoforms of DGK have been identified, and we show that DGKζ is the most abundant isoform expressed by human melanocytic cells. Melanin content, tyrosinase activity, and tyrosinase protein levels were significantly reduced by a DGK inhibitor, but tyrosinase and microphthalmia-associated transcription factor messenger RNA (mRNA) levels were not changed by that inhibition, and there were no effects on the expression of other melanogenesis-related proteins. Isoform-specific small interfering RNAs showed that knockdown of DGKζ decreased melanin content and tyrosinase expression in melanocytic cells. Overexpression of DGKζ increased tyrosinase protein levels, but did not increase tyrosinase mRNA levels. Glycosidase digestion revealed that inhibition of DGK reduced only the mature form of tyrosinase, and the decrease of tyrosinase resulting from DGK inhibition could be blocked partially by protease inhibitors. These results suggest that DGK regulates melanogenesis via modulation of the posttranslational processing of tyrosinase, which may be related with the protein degradation machinery.


Assuntos
Diacilglicerol Quinase/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Melanócitos/enzimologia , Monofenol Mono-Oxigenase/genética , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Diacilglicerol Quinase/antagonistas & inibidores , Diacilglicerol Quinase/genética , Retículo Endoplasmático/enzimologia , Inibidores Enzimáticos/farmacologia , Células Epidérmicas , Flavonoides/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hormônios/metabolismo , Hormônios/farmacologia , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Melaninas/biossíntese , Melaninas/metabolismo , Melanócitos/citologia , Melanoma Experimental , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Oxirredutases/genética , Oxirredutases/metabolismo , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/fisiologia , Quinazolinonas/farmacologia , Neoplasias Cutâneas , alfa-MSH/metabolismo , alfa-MSH/farmacologia
10.
Proc Natl Acad Sci U S A ; 108(16): 6597-602, 2011 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-21460252

RESUMO

The identification of genes that participate in melanomagenesis should suggest strategies for developing therapeutic modalities. We used a public array comparative genomic hybridization (CGH) database and real-time quantitative PCR (qPCR) analyses to identify the AMP kinase (AMPK)-related kinase NUAK2 as a candidate gene for melanomagenesis, and we analyzed its functions in melanoma cells. Our analyses had identified a locus at 1q32 where genomic gain is strongly associated with tumor thickness, and we used real-time qPCR analyses and regression analyses to identify NUAK2 as a candidate gene at that locus. Associations of relapse-free survival and overall survival of 92 primary melanoma patients with NUAK2 expression measured using immunohistochemistry were investigated using Kaplan-Meier curves, log rank tests, and Cox regression models. Knockdown of NUAK2 induces senescence and reduces S-phase, decreases migration, and down-regulates expression of mammalian target of rapamycin (mTOR). In vivo analysis demonstrated that knockdown of NUAK2 suppresses melanoma tumor growth in mice. Survival analysis showed that the risk of relapse is greater in acral melanoma patients with high levels of NUAK2 expression than in acral melanoma patients with low levels of NUAK2 expression (hazard ratio = 3.88; 95% confidence interval = 1.44-10.50; P = 0.0075). These data demonstrate that NUAK2 expression is significantly associated with the oncogenic features of melanoma cells and with the survival of acral melanoma patients. NUAK2 may provide a drug target to suppress melanoma progression. This study further supports the importance of NUAK2 in cancer development and tumor progression, while AMPK has antioncogenic properties.


Assuntos
Movimento Celular , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Melanoma/enzimologia , Melanoma/mortalidade , Proteínas de Neoplasias/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Animais , Senescência Celular/genética , Intervalo Livre de Doença , Feminino , Técnicas de Silenciamento de Genes , Loci Gênicos/genética , Estudo de Associação Genômica Ampla , Humanos , Masculino , Melanoma/genética , Melanoma/patologia , Melanoma/terapia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinases/genética , Fase S/genética , Taxa de Sobrevida , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
11.
Pigment Cell Melanoma Res ; 22(6): 740-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19725928

RESUMO

Because melanomas are intrinsically resistant to conventional radiotherapy and chemotherapy, many alternative treatment approaches have been developed such as biochemotherapy and immunotherapy. The most common cause of multidrug resistance (MDR) in human cancers is the expression and function of one or more ATP-binding cassette (ABC) transporters that efflux anticancer drugs from cells. Melanoma cells express a group of ABC transporters (such as ABCA9, ABCB1, ABCB5, ABCB8, ABCC1, ABCC2, and ABCD1) that may be associated with the resistance of melanoma cells to a broad range of anticancer drugs and/or of melanocytes to toxic melanin intermediates and metabolites. In this review, we propose a model (termed the ABC-M model) in which the intrinsic MDR of melanoma cells is at least in part because of the transporter systems that may also play a critical role in reducing the cytotoxicity of the melanogenic pathway in melanocytes. The ABC-M model suggests molecular strategies to reverse MDR function in the context of the melanogenic pathway, which could open therapeutic avenues towards the ultimate goal of circumventing clinical MDR in patients with melanoma.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Resistência a Múltiplos Medicamentos/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Melaninas/biossíntese , Melanoma/patologia , Melanoma/fisiopatologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Humanos , Melanócitos/citologia , Melanócitos/fisiologia , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Melanossomas/metabolismo , Modelos Biológicos , Proteína 2 Associada à Farmacorresistência Múltipla , Metástase Neoplásica , Células-Tronco Neoplásicas/metabolismo
12.
J Natl Cancer Inst ; 101(18): 1259-71, 2009 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-19704071

RESUMO

BACKGROUND: Malignant melanomas are intrinsically resistant to many conventional treatments, such as radiation and chemotherapy, for reasons that are poorly understood. Here we propose and test a model that explains drug resistance or sensitivity in terms of melanosome dynamics. METHODS: The growth and sensitivity to cisplatin of MNT-1 cells, which are melanotic and enriched with mature stage III and IV melanosomes, and SK-MEL-28 cells, which have only immature stage I and II melanosomes, were compared using clonogenic assays. Differences in pigmentation, melanosome stages, melanosome number, and cellular structures in different cell lines in response to various treatments were examined by electron microscopy. The relative numbers of melanosomes of different stages were compared after treatment with 1-phenyl-2-thiourea. The relationship between drug transporter function and endogenous melanogenic toxicity was assessed by treating cells with the cyclosporin analog PSC-833 and by assessing vacuole formation and cell growth inhibition. All statistical tests were two-sided. RESULTS: Endogenous melanogenic cytotoxicity, produced by damaged melanosomes, resulted in pronounced cell growth inhibition in MNT-1 cells compared with amelanotic SK-MEL-28 cells. The sensitivity to CDDP of MNT-1 cells was 3.8-fold higher than that of SK-MEL-28 cells (mean IC(50) for SK-MEL-28 and MNT-1 = 2.13 microM and 0.56 microM, respectively; difference = 1.57 microM, 95% confidence interval = 1.45 to 1.69; P = .0017). After treatment with 6.7 microM CDDP for 72 hours, the number of stage II-III melanosomes in surviving MNT-1 cells was 6.8-fold that of untreated cells. Modulation of MNT-1 cells to earlier-stage (II, II-III, III) melanosomes by treatment with the tyrosinase inhibitor 1-phenyl-2-thiourea dramatically increased CDDP resistance. Furthermore, PSC-833 principally suppressed MNT-1 melanotic cell growth via an elevation of autophagosome-like vacuolar structures, possibly by inhibiting melanosome membrane transporters. CONCLUSIONS: Melanosome dynamics (including their biogenesis, density, status, and structural integrity) regulate the drug resistance of melanoma cells. Manipulation of melanosome functions may be an effective way to enhance the therapeutic activity of anticancer drugs against melanoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistencia a Medicamentos Antineoplásicos , Melanoma/tratamento farmacológico , Melanoma/patologia , Melanossomas/efeitos dos fármacos , Melanossomas/ultraestrutura , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Ciclosporinas/farmacologia , Doxorrubicina/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Microscopia Confocal , Microscopia Eletrônica , Verapamil/farmacologia , Vimblastina/farmacologia
13.
J Cell Physiol ; 220(3): 640-54, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19452503

RESUMO

The melanocortin 1 receptor (MC1R), a Gs protein-coupled receptor (GPCR) expressed in melanocytes, is a major determinant of skin pigmentation and phototype. MC1R activation stimulates melanogenesis and increases the ratio of black, strongly photoprotective eumelanins to reddish, poorly photoprotective pheomelanins. Several MC1R alleles are associated with red hair, fair skin, increased sensitivity to ultraviolet radiation (the RHC phenotype) and increased skin cancer risk. Three highly penetrant RHC variants, R151C, R160W, and D294H are loss-of-function MC1R mutants with altered cell surface expression. In this study, we show that forward trafficking was normal for D294H. Conversely, export traffic was impaired for R151C, which accumulated in the endoplasmic reticulum (ER), and for R160W, which was enriched in the cis-Golgi. This is the first report of steady-state retention in a post-ER secretory compartment of a GPCR mutant found in the human population. Residues R151 and R160 are located in the MC1R second intracellular loop (il2). Two other mutations in il2, T157A preventing T157 phosphorylation and R162P disrupting a (160)RARR(163) motif, also caused intracellular retention. Moreover, T157 was phosphorylated in wild-type MC1R and a T157D mutation mimicking constitutive phosphorylation allowed normal traffic, and rescued the retention phenotype of R160W and R162P. Therefore, MC1R export is likely regulated by T157 phosphorylation and the (160)RARR(163) arginine-based motif functions as an ER retrieval signal. These elements are conserved in mammalian MC1Rs and in all five types of human melanocortin receptors. Thus, members of this GPCR subfamily might share common mechanisms for regulation of plasma membrane expression.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Cor de Cabelo , Melanócitos/metabolismo , Receptor Tipo 1 de Melanocortina/metabolismo , Neoplasias Cutâneas/metabolismo , Pigmentação da Pele , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular , Humanos , Dados de Sequência Molecular , Mutação , Fosforilação , Conformação Proteica , Transporte Proteico , Receptor Tipo 1 de Melanocortina/genética , Neoplasias Cutâneas/genética , Relação Estrutura-Atividade , Fatores de Tempo , Transfecção
14.
J Clin Invest ; 119(4): 954-63, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19273910

RESUMO

Treatments for primary and metastatic melanomas are rarely effective. Even therapeutics such as retinoic acid (RA) that are successfully used to treat several other forms of cancer are ineffective. Recent evidence indicates that the antiproliferative effects of RA are mediated by the transcription factor SOX9 in human cancer cell lines. As we have previously shown that SOX9 is expressed in normal melanocytes, here we investigated SOX9 expression and function in human melanomas. Although SOX9 was expressed in normal human skin, it was increasingly downregulated as melanocytes progressed to the premalignant and then the malignant and metastatic states. Overexpression of SOX9 in both human and mouse melanoma cell lines induced cell cycle arrest by increasing p21 transcription and restored sensitivity to RA by downregulating expression of PRAME, a melanoma antigen. Furthermore, SOX9 overexpression in melanoma cell lines inhibited tumorigenicity both in mice and in a human ex vivo model of melanoma. Treatment of melanoma cell lines with PGD2 increased SOX9 expression and restored sensitivity to RA. Thus, combined treatment with PGD2 and RA substantially decreased tumor growth in human ex vivo and mouse in vivo models of melanoma. The results of our experiments targeting SOX9 provide insight into the pathophysiology of melanoma. Further, the effects of SOX9 on melanoma cell proliferation and RA sensitivity suggest the encouraging possibility of a noncytotoxic approach to the treatment of melanoma.


Assuntos
Melanoma/genética , Melanoma/patologia , Fatores de Transcrição SOX9/genética , Animais , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação para Baixo , Humanos , Melanoma/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Fator de Transcrição Associado à Microftalmia/genética , Nevo/genética , Nevo/patologia , Prostaglandina D2/farmacologia , Fatores de Transcrição SOX9/fisiologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Tretinoína/farmacologia , Regulação para Cima/efeitos dos fármacos
15.
Proc Natl Acad Sci U S A ; 106(6): 1802-7, 2009 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19174519

RESUMO

The melanocortin-1 receptor (MC1R) is a key regulator of pigmentation in mammals and is tightly linked to an increased risk of skin cancers, including melanoma, in humans. Physiologically activated by alpha-melanocyte stimulating hormone (alphaMSH), MC1R function can be antagonized by a secreted factor, agouti signal protein (ASP), which is responsible for the lighter phenotypes in mammals (including humans), and is also associated with increased risk of skin cancer. It is therefore of great interest to characterize the molecular effects elicited by those MC1R ligands. In this study, we determined the gene expression profiles of murine melan-a melanocytes treated with ASP or alphaMSH over a 4-day time course using genome-wide oligonucleotide microarrays. As expected, there were significant reductions in expression of numerous melanogenic proteins elicited by ASP, which correlates with its inhibition of pigmentation. ASP also unexpectedly modulated the expression of genes involved in various other cellular pathways, including glutathione synthesis and redox metabolism. Many genes up-regulated by ASP are involved in morphogenesis (especially in nervous system development), cell adhesion, and extracellular matrix-receptor interactions. Concomitantly, ASP enhanced the migratory potential and the invasiveness of melanocytic cells in vitro. These results demonstrate the role of ASP in the dedifferentiation of melanocytes, identify pigment-related genes targeted by ASP and by alphaMSH, and provide insights into the pleiotropic molecular effects of MC1R signaling that may function during development and may affect skin cancer risk.


Assuntos
Proteína Agouti Sinalizadora/fisiologia , Diferenciação Celular , Perfilação da Expressão Gênica , Melanócitos/citologia , Receptor Tipo 1 de Melanocortina/metabolismo , Proteína Agouti Sinalizadora/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Suscetibilidade a Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Genômica , Ligantes , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Pigmentação/efeitos dos fármacos , Pigmentação/genética , Transdução de Sinais , Neoplasias Cutâneas/etiologia , alfa-MSH/farmacologia
16.
Anal Biochem ; 383(2): 265-9, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18801330

RESUMO

Most current techniques employed to improve antigen-antibody signals in Western blotting and in immunohistochemistry rely on sample processing prior to staining (e.g., microwaving) or using a more robust reporter (e.g., a secondary antibody with biotin-streptavidin). We have developed and optimized a new approach intended to stabilize the complexes formed between antigens and their respective primary antibodies by cupric ions at high pH. This technique improves the affinity and lowers cross-reactivity with nonspecific bands of approximately 20% of antibodies tested (5/25). Here we report that this method can enhance antigen-antibody specificity and can improve the utility of some poorly reactive primary antibodies.


Assuntos
Especificidade de Anticorpos , Cobre/química , Reagentes de Ligações Cruzadas/química , Peptídeos/química , Peptídeos/metabolismo , Animais , Afinidade de Anticorpos/efeitos dos fármacos , Especificidade de Anticorpos/efeitos dos fármacos , Reações Antígeno-Anticorpo/efeitos dos fármacos , Reação de Biureto , Cobre/farmacologia , Reações Cruzadas/efeitos dos fármacos , Reagentes de Ligações Cruzadas/farmacologia , Humanos , Camundongos
17.
J Invest Dermatol ; 128(1): 162-74, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17687388

RESUMO

Melanosomes are unique membrane-bound organelles specialized for the synthesis and distribution of melanin. Mechanisms involved in the trafficking of proteins to melanosomes and in the transport of mature pigmented melanosomes to the dendrites of melanocytic cells are being characterized, but details about those processes during early stages of melanosome maturation are not well understood. Early melanosomes must remain in the perinuclear area until critical components are assembled. In this study, we characterized the processing of two distinct melanosomal proteins, tyrosinase (TYR) and Pmel17, to elucidate protein processing in early or late steps of the secretory pathway, respectively, and to determine mechanisms underlying the subcellular localization and transport of early melanosomes. We used immunological, biochemical, and molecular approaches to demonstrate that the movement of early melanosomes in the perinuclear area depends primarily on microtubules but not on actin filaments. In contrast, the trafficking of TYR and Pmel17 depends on cytoplasmic dynein and its interaction with the spectrin/ankyrin system, which is involved with the sorting of cargo from the plasma membrane. These results provide important clues toward understanding the processes involved with early events in melanosome formation and transport.


Assuntos
Dineínas/fisiologia , Melanossomas/fisiologia , Glicoproteínas de Membrana/metabolismo , Monofenol Mono-Oxigenase/metabolismo , Espectrina/fisiologia , Actinas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteína Agouti Sinalizadora/fisiologia , Animais , Transporte Biológico , Células Cultivadas , Di-Hidroxifenilalanina/análise , Complexo de Golgi/metabolismo , Humanos , Cinesinas/análise , Melanoma/metabolismo , Melanossomas/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Microtúbulos/fisiologia , Transporte Proteico , Espectrina/análise , Antígeno gp100 de Melanoma , Proteínas rab de Ligação ao GTP/análise , Proteínas rab27 de Ligação ao GTP
18.
Proc Natl Acad Sci U S A ; 104(35): 13984-9, 2007 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-17702866

RESUMO

SOX (SRY type HMG box) proteins are transcription factors that are predominantly known for their roles during development. During melanocyte development from the neural crest, SOX10 regulates microphthalmia-associated transcription factor, which controls a set of genes critical for pigment cell development and pigmentation, including dopachrome tautomerase and tyrosinase. We report here that another SOX factor, SOX9, is expressed by melanocytes in neonatal and adult human skin and is up-regulated by UVB exposure. We demonstrate that this regulation is mediated by cAMP and protein kinase. We also show that agouti signal protein, a secreted factor known to decrease pigmentation, down-regulates SOX9 expression. In adult and neonatal melanocytes, SOX9 regulates microphthalmia-associated transcription factor, dopachrome tautomerase, and tyrosinase promoters, leading to an increase in the expression of these key melanogenic proteins and finally to a stimulation of pigmentation. SOX9 completes the complex and tightly regulated process leading to the production of melanin by acting at a very upstream level. This role of SOX9 in pigmentation emphasizes the poorly understood impact of SOX proteins in adult tissues.


Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Proteínas de Grupo de Alta Mobilidade/genética , Melanócitos/citologia , Melanócitos/fisiologia , Pigmentação da Pele/fisiologia , Fatores de Transcrição/genética , Diferenciação Celular , AMP Cíclico/fisiologia , Humanos , Hibridização In Situ , Melanócitos/efeitos da radiação , Proteínas Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9 , Raios Ultravioleta
19.
J Biol Chem ; 282(15): 11266-80, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17303571

RESUMO

Pmel17 is a melanocyte/melanoma-specific protein that is essential for the maturation of melanosomes to form mature, fibrillar, and pigmented organelles. Recently, we reported that the less glycosylated form of Pmel17 (termed iPmel17) is sorted via the plasma membrane in a manner distinct from mature Pmel17 (termed mPmel17), which is sorted directly to melanosomes. To clarify the mechanism(s) underlying the distinct processing and sorting of Pmel17, we generated a highly specific antibody (termed alphaPEP25h) against an epitope within the repeat domain of Pmel17 that is sensitive to changes in O-glycosylation. alphaPEP25h recognizes only iPmel17 and allows analysis of the processing and sorting of iPmel17 when compared with alphaPEP13h, an antibody that recognizes both iPmel17 and mPmel17. Our novel findings using alphaPEP25h demonstrate that iPmel17 differs from mPmel17 not only in its sensitivity to endoglycosidase H, but also in the content of core 1 O-glycans modified with sialic acid. This evidence reveals that iPmel17 is glycosylated differently in the Golgi and that it is sorted through the secretory pathway. Analysis of Pmel17 processing in glycosylation-deficient mutant cells reveals that Pmel17 lacking the correct addition of sialic acid and galactose loses the ability to form fibrils. Furthermore, we show that addition of sialic acid affects the stability and sorting of Pmel17 and reduces pigmentation. Alterations in sialyltransferase activity and substrates differ between normal and transformed melanocytes and may represent a critical change during malignant transformation.


Assuntos
Glicoproteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Animais , Anticorpos/imunologia , Linhagem Celular Tumoral , Cricetinae , Retículo Endoplasmático/metabolismo , Humanos , Melanossomas/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Microscopia Imunoeletrônica , Ácido N-Acetilneuramínico/metabolismo , Transporte Proteico , Fatores de Tempo , Antígeno gp100 de Melanoma
20.
J Proteome Res ; 5(11): 3135-44, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17081065

RESUMO

Melanin, which is responsible for virtually all visible skin, hair, and eye pigmentation in humans, is synthesized, deposited, and distributed in subcellular organelles termed melanosomes. A comprehensive determination of the protein composition of this organelle has been obstructed by the melanin present. Here, we report a novel method of removing melanin that includes in-solution digestion and immobilized metal affinity chromatography (IMAC). Together with in-gel digestion, this method has allowed us to characterize melanosome proteomes at various developmental stages by tandem mass spectrometry. Comparative profiling and functional characterization of the melanosome proteomes identified approximately 1500 proteins in melanosomes of all stages, with approximately 600 in any given stage. These proteins include 16 homologous to mouse coat color genes and many associated with human pigmentary diseases. Approximately 100 proteins shared by melanosomes from pigmented and nonpigmented melanocytes define the essential melanosome proteome. Proteins validated by confirming their intracellular localization include PEDF (pigment-epithelium derived factor) and SLC24A5 (sodium/potassium/calcium exchanger 5, NCKX5). The sharing of proteins between melanosomes and other lysosome-related organelles suggests a common evolutionary origin. This work represents a model for the study of the biogenesis of lysosome-related organelles.


Assuntos
Melanossomas/fisiologia , Proteômica/métodos , Animais , Linhagem Celular Tumoral , Cromatografia de Afinidade , Biologia Computacional/métodos , Cor de Olho , Perfilação da Expressão Gênica , Cor de Cabelo , Humanos , Melanoma , Melanossomas/química , Camundongos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/isolamento & purificação , Biogênese de Organelas , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/isolamento & purificação , Reprodutibilidade dos Testes , Tripsina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA