Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Commun Biol ; 4(1): 1412, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34921240

RESUMO

Effective conservation and management of threatened wildlife populations require an accurate assessment of age structure to estimate demographic trends and population viability. Epigenetic aging models are promising developments because they estimate individual age with high accuracy, accurately predict age in related species, and do not require invasive sampling or intensive long-term studies. Using blood and biopsy samples from known age plains zebras (Equus quagga), we model epigenetic aging using two approaches: the epigenetic clock (EC) and the epigenetic pacemaker (EPM). The plains zebra EC has the potential for broad application within the genus Equus given that five of the seven extant wild species of the genus are threatened. We test the EC's ability to predict age in sister taxa, including two endangered species and the more distantly related domestic horse, demonstrating high accuracy in all cases. By comparing chronological and estimated age in plains zebras, we investigate age acceleration as a proxy of health status. An interaction between chronological age and inbreeding is associated with age acceleration estimated by the EPM, suggesting a cumulative effect of inbreeding on biological aging throughout life.


Assuntos
Distribuição por Idade , Epigênese Genética , Equidae/genética , Animais , Espécies em Perigo de Extinção , Epigenômica , Equidae/fisiologia , Cavalos/fisiologia , Modelos Genéticos , Dinâmica Populacional , Especificidade da Espécie
2.
PLoS Genet ; 14(11): e1007671, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30500825

RESUMO

Mutations that alter signaling of RAS/MAPK-family proteins give rise to a group of Mendelian diseases known as RASopathies. However, among RASopathies, the matrix of genotype-phenotype relationships is still incomplete, in part because there are many RAS-related proteins and in part because the phenotypic consequences may be variable and/or pleiotropic. Here, we describe a cohort of ten cases, drawn from six clinical sites and over 16,000 sequenced probands, with de novo protein-altering variation in RALA, a RAS-like small GTPase. All probands present with speech and motor delays, and most have intellectual disability, low weight, short stature, and facial dysmorphism. The observed rate of de novo RALA variants in affected probands is significantly higher (p = 4.93 x 10(-11)) than expected from the estimated random mutation rate. Further, all de novo variants described here affect residues within the GTP/GDP-binding region of RALA; in fact, six alleles arose at only two codons, Val25 and Lys128. The affected residues are highly conserved across both RAL- and RAS-family genes, are devoid of variation in large human population datasets, and several are homologous to positions at which disease-associated variants have been observed in other GTPase genes. We directly assayed GTP hydrolysis and RALA effector-protein binding of the observed variants, and found that all but one tested variant significantly reduced both activities compared to wild-type. The one exception, S157A, reduced GTP hydrolysis but significantly increased RALA-effector binding, an observation similar to that seen for oncogenic RAS variants. These results show the power of data sharing for the interpretation and analysis of rare variation, expand the spectrum of molecular causes of developmental disability to include RALA, and provide additional insight into the pathogenesis of human disease caused by mutations in small GTPases.


Assuntos
Deficiências do Desenvolvimento/genética , Deficiência Intelectual/genética , Proteínas Mitocondriais/genética , Mutação , Domínios e Motivos de Interação entre Proteínas/genética , Proteínas ral de Ligação ao GTP/genética , Proteínas ras/genética , Fácies , Genótipo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Proteínas Mitocondriais/química , Modelos Moleculares , Mutação de Sentido Incorreto , Fenótipo , Conformação Proteica , Proteínas ral de Ligação ao GTP/química , Proteínas ras/química
3.
Biophys J ; 109(9): 1946-58, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26536271

RESUMO

The ß-defensins are a class of small cationic proteins that serve as components of numerous systems in vertebrate biology, including the immune and melanocortin systems. Human ß-defensin 3 (HBD3), which is produced in the skin, has been found to bind to melanocortin receptors 1 and 4 through complementary electrostatics, a unique mechanism of ligand-receptor interaction. This finding indicates that electrostatics alone, and not specific amino acid contact points, could be sufficient for function in this ligand-receptor system, and further suggests that other small peptide ligands could interact with these receptors in a similar fashion. Here, we conducted molecular-similarity analyses and functional studies of additional members of the human ß-defensin family, examining their potential as ligands of melanocortin-1 receptor, through selection based on their electrostatic similarity to HBD3. Using Poisson-Boltzmann electrostatic calculations and molecular-similarity analysis, we identified members of the human ß-defensin family that are both similar and dissimilar to HBD3 in terms of electrostatic potential. Synthesis and functional testing of a subset of these ß-defensins showed that peptides with an HBD3-like electrostatic character bound to melanocortin receptors with high affinity, whereas those that were anticorrelated to HBD3 showed no binding affinity. These findings expand on the central role of electrostatics in the control of this ligand-receptor system and further demonstrate the utility of employing molecular-similarity analysis. Additionally, we identified several new potential ligands of melanocortin-1 receptor, which may have implications for our understanding of the role defensins play in melanocortin physiology.


Assuntos
Receptor Tipo 1 de Melanocortina/química , Eletricidade Estática , beta-Defensinas/química , Sequência de Aminoácidos , Ligação Competitiva , Bases de Dados de Proteínas , Humanos , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica , Dobramento de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , beta-Defensinas/genética
4.
PLoS One ; 10(6): e0128969, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26042826

RESUMO

Coat color in Holstein dairy cattle is primarily controlled by the melanocortin 1 receptor (MC1R) gene, a central determinant of black (eumelanin) vs. red/brown pheomelanin synthesis across animal species. The major MC1R alleles in Holsteins are Dominant Black (MC1RD) and Recessive Red (MC1Re). A novel form of dominant red coat color was first observed in an animal born in 1980. The mutation underlying this phenotype was named Dominant Red and is epistatic to the constitutively activated MC1RD. Here we show that a missense mutation in the coatomer protein complex, subunit alpha (COPA), a gene with previously no known role in pigmentation synthesis, is completely associated with Dominant Red in Holstein dairy cattle. The mutation results in an arginine to cysteine substitution at an amino acid residue completely conserved across eukaryotes. Despite this high level of conservation we show that both heterozygotes and homozygotes are healthy and viable. Analysis of hair pigment composition shows that the Dominant Red phenotype is similar to the MC1R Recessive Red phenotype, although less effective at reducing eumelanin synthesis. RNA-seq data similarly show that Dominant Red animals achieve predominantly pheomelanin synthesis by downregulating genes normally required for eumelanin synthesis. COPA is a component of the coat protein I seven subunit complex that is involved with retrograde and cis-Golgi intracellular coated vesicle transport of both protein and RNA cargo. This suggests that Dominant Red may be caused by aberrant MC1R protein or mRNA trafficking within the highly compartmentalized melanocyte, mimicking the effect of the Recessive Red loss of function MC1R allele.


Assuntos
Proteína Coatomer/genética , Genes Dominantes , Estudos de Associação Genética , Mutação de Sentido Incorreto/genética , Pigmentação da Pele/genética , Animais , Bovinos/genética , Mapeamento Cromossômico , Sequência Conservada/genética , Evolução Molecular , Genoma , Cor de Cabelo/genética , Fenótipo , Receptor Tipo 1 de Melanocortina/genética , Reprodutibilidade dos Testes , Análise de Sequência de DNA , Análise de Sequência de RNA
5.
Pigment Cell Melanoma Res ; 25(4): 411-33, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22578174

RESUMO

Animals display incredibly diverse color patterns yet little is known about the underlying genetic basis of these phenotypes. However, emerging results are reshaping our view of how the process of phenotypic evolution occurs. Here, we outline recent research from three particularly active areas of investigation: melanin pigmentation in Drosophila, wing patterning in butterflies, and pigment variation in lizards. For each system, we highlight (i) the function and evolution of color variation, (ii) various approaches that have been used to explore the genetic basis of pigment variation, and (iii) conclusions regarding the genetic basis of convergent evolution which have emerged from comparative analyses. Results from these studies indicate that natural variation in pigmentation is a particularly powerful tool to examine the molecular basis of evolution, especially with regard to convergent or parallel evolution. Comparison of these systems also reveals that the molecular basis of convergent evolution is heterogeneous, sometimes involving conserved mechanisms and sometimes not. In the near future, additional work in other emerging systems will substantially expand the scope of available comparisons.


Assuntos
Variação Genética , Natureza , Pigmentação/genética , Animais , Borboletas/anatomia & histologia , Borboletas/genética , Melaninas/metabolismo , Asas de Animais/anatomia & histologia
6.
Genome Res ; 21(11): 1905-15, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21844123

RESUMO

Next-generation sequencing technologies offer new approaches for global measurements of gene expression but are mostly limited to organisms for which a high-quality assembled reference genome sequence is available. We present a method for gene expression profiling called EDGE, or EcoP15I-tagged Digital Gene Expression, based on ultra-high-throughput sequencing of 27-bp cDNA fragments that uniquely tag the corresponding gene, thereby allowing direct quantification of transcript abundance. We show that EDGE is capable of assaying for expression in >99% of genes in the genome and achieves saturation after 6-8 million reads. EDGE exhibits very little technical noise, reveals a large (10(6)) dynamic range of gene expression, and is particularly suited for quantification of transcript abundance in non-model organisms where a high-quality annotated genome is not available. In a direct comparison with RNA-seq, both methods provide similar assessments of relative transcript abundance, but EDGE does better at detecting gene expression differences for poorly expressed genes and does not exhibit transcript length bias. Applying EDGE to laboratory mice, we show that a loss-of-function mutation in the melanocortin 1 receptor (Mc1r), recognized as a Mendelian determinant of yellow hair color in many different mammals, also causes reduced expression of genes involved in the interferon response. To illustrate the application of EDGE to a non-model organism, we examine skin biopsy samples from a cheetah (Acinonyx jubatus) and identify genes likely to control differences in the color of spotted versus non-spotted regions.


Assuntos
Perfilação da Expressão Gênica , Expressão Gênica , Acinonyx/genética , Animais , Biologia Computacional , Etiquetas de Sequências Expressas , Regulação da Expressão Gênica/efeitos dos fármacos , Interferons/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Pigmentação/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transcrição Gênica/efeitos dos fármacos
7.
Blood ; 118(13): 3622-33, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21788341

RESUMO

Reduced gene dosage of ribosomal protein subunits has been implicated in 5q- myelodysplastic syndrome and Diamond Blackfan anemia, but the cellular and pathophysiologic defects associated with these conditions are enigmatic. Using conditional inactivation of the ribosomal protein S6 gene in laboratory mice, we found that reduced ribosomal protein gene dosage recapitulates cardinal features of the 5q- syndrome, including macrocytic anemia, erythroid hypoplasia, and megakaryocytic dysplasia with thrombocytosis, and that p53 plays a critical role in manifestation of these phenotypes. The blood cell abnormalities are accompanied by a reduction in the number of HSCs, a specific defect in late erythrocyte development, and suggest a disease-specific ontogenetic pathway for megakaryocyte development. Further studies of highly purified HSCs from healthy patients and from those with myelodysplastic syndrome link reduced expression of ribosomal protein genes to decreased RBC maturation and suggest an underlying and common pathophysiologic pathway for additional subtypes of myelodysplastic syndrome.


Assuntos
Dosagem de Genes , Síndromes Mielodisplásicas/genética , Proteínas Ribossômicas/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Regulação para Baixo/genética , Feminino , Dosagem de Genes/genética , Dosagem de Genes/fisiologia , Predisposição Genética para Doença , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/metabolismo , Proteínas Ribossômicas/metabolismo , Fatores de Risco , Proteína Supressora de Tumor p53/fisiologia
8.
PLoS One ; 6(4): e18324, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21694754

RESUMO

Insulin and leptin intracellular signaling pathways converge and act synergistically on the hypothalamic phosphatidylinositol-3-OH kinase/3-phosphoinositide-dependent protein kinase 1 (PDK1). However, little is known about whether PDK1 in agouti-related peptide (AGRP) neurons contributes to energy homeostasis. We generated AGRP neuron-specific PDK1 knockout (AGRPPdk1(-/-)) mice and mice with selective expression of transactivation-defective Foxo1 (Δ256Foxo1(AGRP)Pdk1(-/-)). The AGRPPdk1(-/-) mice showed reductions in food intake, body length, and body weight. The Δ256Foxo1(AGRP)Pdk1(-/-) mice showed increased body weight, food intake, and reduced locomotor activity. After four weeks of calorie-restricted feeding, oxygen consumption and locomotor activity were elevated in AGRPPdk1(-/-) mice and reduced in Δ256Foxo1(AGRP)Pdk1(-/-) mice. In vitro, ghrelin-induced changes in [Ca(2+)](i) and inhibition of ghrelin by leptin were significantly attenuated in AGRPPdk1(-/-) neurons compared to control neurons. However, ghrelin-induced [Ca(2+)](i) changes and leptin inhibition were restored in Δ256Foxo1(AGRP)Pdk1(-/-) mice. These results suggested that PDK1 and Foxo1 signaling pathways play important roles in the control of energy homeostasis through AGRP-independent mechanisms.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Ingestão de Alimentos , Metabolismo Energético , Fatores de Transcrição Forkhead/metabolismo , Melanocortinas/metabolismo , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Cálcio/metabolismo , Restrição Calórica , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Feminino , Proteína Forkhead Box O1 , Técnicas de Inativação de Genes , Grelina/farmacologia , Homeostase/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Fenótipo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
9.
J Mol Biol ; 404(1): 45-55, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-20831872

RESUMO

Agouti-related protein (AgRP) and agouti signaling protein (ASIP) are homologs that play critical roles in energy balance and pigmentation, respectively, by functioning as antagonistic ligands at their cognate melanocortin receptors. Signaling specificity is mediated in part through receptor binding selectivity brought about by alterations in the cysteine-rich carboxy-terminal domains of the ligands. AgRP binds with high affinity to the melanocortin 3 receptor and the melanocortin 4 receptor, but not to the melanocortin 1 receptor (MC1R), whereas ASIP binds with high affinity to all three receptors. This work explores the structural basis for receptor selectivity by studying chimeric proteins developed by interchanging loops between the cysteine-rich domain of ASIP and the cysteine-rich domain of AgRP. Binding data demonstrate that melanocortin 4 receptor responds to all chimeras and is therefore highly tolerant of gross loop changes. By contrast, MC1R responds primarily to those chimeras with a sequence close to that of wild-type ASIP. Further analysis of binding and functional data suggests that the ASIP C-terminal loop (a six-amino-acid segment closed by the final disulfide bond) is essential for high-affinity MC1R binding and inverse agonism. Comparison with previously published molecular models suggests that this loop makes contact with the first extracellular loop of MC1R through a series of key hydrophobic interactions.


Assuntos
Proteína Agouti Sinalizadora/genética , Proteína Agouti Sinalizadora/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Mapeamento de Interação de Proteínas , Receptor Tipo 1 de Melanocortina/antagonistas & inibidores , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Receptor Tipo 3 de Melanocortina/antagonistas & inibidores , Receptor Tipo 4 de Melanocortina/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
Pigment Cell Melanoma Res ; 22(6): 819-26, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19627560

RESUMO

Hair color and skin color are frequently coordinated in mammalian species. To explore this, we have studied mutations in two different G protein coupled pathways, each of which affects the darkness of both hair and skin color. In each mouse mutant (Gnaq(Dsk1), Gna11(Dsk7), and Mc1r(e)), we analyzed the melanocyte density and the concentrations of eumelanin (black pigment) and pheomelanin (yellow pigment) in the hair or skin to determine the mechanisms regulating pigmentation. Surprisingly, we discovered that each mutation affects hair and skin color differently. Furthermore, we have found that in the epidermis, the melanocortin signaling pathway does not couple the synthesis of eumelanin with pheomelanin, as it does in hair follicles. Even by shared signaling pathways, hair and skin melanocytes are regulated quite independently.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Cor de Cabelo/fisiologia , Pigmentação/fisiologia , Transdução de Sinais/fisiologia , Pigmentação da Pele/fisiologia , Animais , Endotelinas/metabolismo , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Humanos , Masculino , Melaninas/metabolismo , Melanócitos/citologia , Melanócitos/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/metabolismo , Receptor Tipo 1 de Melanocortina/metabolismo
11.
Pigment Cell Melanoma Res ; 22(5): 623-34, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19493315

RESUMO

Melanocortin-1 receptor (MC1R) and its ligands, alpha-melanocyte stimulating hormone (alphaMSH) and agouti signaling protein (ASIP), regulate switching between eumelanin and pheomelanin synthesis in melanocytes. Here we investigated biological effects and signaling pathways of ASIP. Melan-a non agouti (a/a) mouse melanocytes produce mainly eumelanin, but ASIP combined with phenylthiourea and extra cysteine could induce over 200-fold increases in the pheomelanin to eumelanin ratio, and a tan-yellow color in pelletted cells. Moreover, ASIP-treated cells showed reduced proliferation and a melanoblast-like appearance, seen also in melanocyte lines from yellow (A(y)/a and Mc1r(e)/ Mc1r(e)) mice. However ASIP-YY, a C-terminal fragment of ASIP, induced neither biological nor pigmentary changes. As, like ASIP, ASIP-YY inhibited the cAMP rise induced by alphaMSH analog NDP-MSH, and reduced cAMP level without added MSH, the morphological changes and depigmentation seemed independent of cAMP signaling. Melanocytes genetically null for ASIP mediators attractin or mahogunin (Atrn(mg-3J/mg-3J) or Mgrn1(md-nc/md-nc)) also responded to both ASIP and ASIP-YY in cAMP level, while only ASIP altered their proliferation and (in part) shape. Thus, ASIP-MC1R signaling includes a cAMP-independent pathway through attractin and mahogunin, while the known cAMP-dependent component requires neither attractin nor mahogunin.


Assuntos
Proteína Agouti Sinalizadora/metabolismo , AMP Cíclico/metabolismo , Melaninas/biossíntese , Melanócitos/fisiologia , Proteínas de Membrana/metabolismo , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Proteína Agouti Sinalizadora/genética , Animais , Proliferação de Células , Forma Celular , Células Cultivadas , Cor de Cabelo/fisiologia , Melaninas/metabolismo , Melanócitos/citologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pigmentação/fisiologia , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 1 de Melanocortina/metabolismo , Ubiquitina-Proteína Ligases/genética
12.
Genome Res ; 19(5): 826-37, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19307593

RESUMO

Genome-wide scans for recent positive selection in humans have yielded insight into the mechanisms underlying the extensive phenotypic diversity in our species, but have focused on a limited number of populations. Here, we present an analysis of recent selection in a global sample of 53 populations, using genotype data from the Human Genome Diversity-CEPH Panel. We refine the geographic distributions of known selective sweeps, and find extensive overlap between these distributions for populations in the same continental region but limited overlap between populations outside these groupings. We present several examples of previously unrecognized candidate targets of selection, including signals at a number of genes in the NRG-ERBB4 developmental pathway in non-African populations. Analysis of recently identified genes involved in complex diseases suggests that there has been selection on loci involved in susceptibility to type II diabetes. Finally, we search for local adaptation between geographically close populations, and highlight several examples.


Assuntos
Genética Populacional , Seleção Genética , Diabetes Mellitus Tipo 2/genética , Receptores ErbB/genética , Predisposição Genética para Doença/genética , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Neurregulinas/genética , Fenótipo , Receptor ErbB-4
13.
Nature ; 457(7229): 599-602, 2009 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-19078957

RESUMO

BRAF and NRAS are common targets for somatic mutations in benign and malignant neoplasms that arise from melanocytes situated in epithelial structures, and lead to constitutive activation of the mitogen-activated protein (MAP) kinase pathway. However, BRAF and NRAS mutations are absent in a number of other melanocytic neoplasms in which the equivalent oncogenic events are currently unknown. Here we report frequent somatic mutations in the heterotrimeric G protein alpha-subunit, GNAQ, in blue naevi (83%) and ocular melanoma of the uvea (46%). The mutations occur exclusively in codon 209 in the Ras-like domain and result in constitutive activation, turning GNAQ into a dominant acting oncogene. Our results demonstrate an alternative route to MAP kinase activation in melanocytic neoplasia, providing new opportunities for therapeutic intervention.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/genética , Melanoma/genética , Mutação/genética , Nevo Azul/genética , Neoplasias Cutâneas/genética , Neoplasias Uveais/genética , Apoptose , Biópsia , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Códon/genética , Análise Mutacional de DNA , Ativação Enzimática , Subunidades alfa de Proteínas de Ligação ao GTP/química , Subunidades alfa de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Genes Dominantes/genética , Humanos , Sistema de Sinalização das MAP Quinases , Melanócitos/enzimologia , Melanócitos/patologia , Melanoma/enzimologia , Melanoma/patologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Nevo Azul/enzimologia , Nevo Azul/patologia , Oncogenes/genética , Estrutura Terciária de Proteína , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Neoplasias Uveais/enzimologia , Neoplasias Uveais/patologia , Proteínas ras/química
14.
Nat Genet ; 40(8): 963-70, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18641651

RESUMO

Mutations in genes encoding ribosomal proteins cause the Minute phenotype in Drosophila and mice, and Diamond-Blackfan syndrome in humans. Here we report two mouse dark skin (Dsk) loci caused by mutations in Rps19 (ribosomal protein S19) and Rps20 (ribosomal protein S20). We identify a common pathophysiologic program in which p53 stabilization stimulates Kit ligand expression, and, consequently, epidermal melanocytosis via a paracrine mechanism. Accumulation of p53 also causes reduced body size and erythrocyte count. These results provide a mechanistic explanation for the diverse collection of phenotypes that accompany reduced dosage of genes encoding ribosomal proteins, and have implications for understanding normal human variation and human disease.


Assuntos
Proteínas Ribossômicas/genética , Pigmentação da Pele , Proteína Supressora de Tumor p53/metabolismo , Animais , Células Epidérmicas , Epiderme/metabolismo , Eritrócitos/metabolismo , Humanos , Queratinócitos/metabolismo , Melanócitos/metabolismo , Camundongos , Mutação , Proteína S6 Ribossômica/genética , Proteína S6 Ribossômica/metabolismo , Proteínas Ribossômicas/metabolismo , Fator de Células-Tronco/metabolismo
15.
Cell Metab ; 7(3): 236-48, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18316029

RESUMO

Leptin, an adipocyte-derived hormone, acts on hypothalamic neurons located in the arcuate nucleus (ARC) of the hypothalamus to regulate energy homeostasis. One of the leptin-regulated neuronal subtypes in the ARC are agouti-related peptide (AgRP)-expressing neurons, which are involved in the regulation of food intake and are directly inhibited by leptin. Leptin activates the signal transducer and activator of transcription 3 (Stat3), but the role of Stat3 in the regulation of AgRP neurons is unclear. Here we show that mice expressing a constitutively active version of Stat3 selectively in AgRP neurons are lean and exhibit relative resistance to diet-induced obesity. Surprisingly, this phenotype arises from increased locomotor activity in the presence of unaltered AgRP expression. These data demonstrate that Stat3-dependent signaling in AgRP neurons in the ARC controls locomotor activity independently of AgRP regulation.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Atividade Motora , Neurônios/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Peso Corporal , Ritmo Circadiano , Gorduras na Dieta/administração & dosagem , Modelos Animais de Doenças , Ingestão de Alimentos , Metabolismo Energético , Feminino , Genótipo , Glucose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/fisiopatologia , Obesidade/prevenção & controle , Fenótipo , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT3/genética , Magreza/metabolismo , Magreza/fisiopatologia
16.
Pigment Cell Melanoma Res ; 21(1): 63-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18353144

RESUMO

Manipulation of gene expression in melanocytes is an important tool for studying pigment cell biology. We constructed transgenic mice in which Cre recombinase was placed under the control of regulatory elements from the Microphthalmia-associated transcriptional factor (Mitf) gene using bacterial artificial chromosome (BAC). Bacterial artificial chromosome that contained either 50 or 108 kb DNA 5' to the melanocyte-specific (1M) transcriptional start site gave rise to transgenic lines in which Cre is expressed specifically in cells of the melanocyte lineage, as judged by activation of the Gt(Rosa)26(tm1Sor)(R26R) reporter locus. Activation of R26R is first detectable in melanoblasts of midgestation embryos, and completely marks all melanocyte components of the skin in postnatal animals. To test the utility of the MitfCre transgene, we used a loxP-targeted allele of the protein kinase A alpha catalytic subunit (Prkaca), modified such that Cre-mediated recombination activates PKA signaling. On an agouti background, animals carrying both the MitfCre transgene and the targeted Prkaca allele (CalphaR) exhibited a darker coat color than control littermates, due to a shift from pheomelanin to eumelanin synthesis. Our results confirm that PKA signaling is a key component of pigment type-switching, and provide a new tool for studying pigment cell biology.


Assuntos
Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Cor de Cabelo/genética , Integrases/genética , Melanócitos/enzimologia , Fator de Transcrição Associado à Microftalmia/genética , Recombinação Genética , Elementos Reguladores de Transcrição , Animais , Cromossomos Artificiais Bacterianos , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Genótipo , Integrases/metabolismo , Melaninas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Fenótipo , Proteínas/genética , Proteínas/metabolismo , RNA não Traduzido , Transdução de Sinais/genética
17.
Mol Cell Biol ; 28(1): 282-92, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17967885

RESUMO

Members of the PIAS (for protein inhibitor of activated STAT) family play critical roles in modulating the activity of a variety of transcriptional regulators. Zimp10, a novel PIAS-like protein, is a transcriptional coregulator and may be involved in the modification of chromatin through interactions with the SWI/SNF chromatin-remodeling complexes. Here, we investigate the biological role of Zimp10 in zimp10-deficient mice. Homozygosity for the Zimp10-targeted allele resulted in developmental arrest at approximately embryonic day 10.5. Analysis of knockout embryos revealed severe defects in the reorganization of the yolk sac vascular plexus. No significant abnormality in hematopoietic potential was observed in zimp10 null mice. Microarray and quantified reverse transcription-PCR analyses showed that the expression of the Fos family member Fra-1, which is involved in extraembryonic vascular development, was reduced in yolk sac tissues of zimp10 null embryos. Using fra-1 promoter/reporter constructs, we further demonstrate the regulatory role of Zimp10 on the transcription of Fra-1. This study provides evidence to demonstrate a crucial role for Zimp10 in vasculogenesis.


Assuntos
Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Perda do Embrião/metabolismo , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/citologia , Feminino , Hematopoese , Homozigoto , Humanos , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Saco Vitelino/irrigação sanguínea , Saco Vitelino/embriologia , Saco Vitelino/metabolismo
18.
Endocrinology ; 149(4): 1773-85, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18162515

RESUMO

Two known types of leptin-responsive neurons reside within the arcuate nucleus: the agouti gene-related peptide (AgRP)/neuropeptide Y (NPY) neuron and the proopiomelanocortin (POMC) neuron. By deleting the leptin receptor gene (Lepr) specifically in AgRP/NPY and/or POMC neurons of mice, we examined the several and combined contributions of these neurons to leptin action. Body weight and adiposity were increased by Lepr deletion from AgRP and POMC neurons individually, and simultaneous deletion in both neurons (A+P LEPR-KO mice) further increased these measures. Young (periweaning) A+P LEPR-KO mice exhibit hyperphagia and decreased energy expenditure, with increased weight gain, oxidative sparing of triglycerides, and increased fat accumulation. Interestingly, however, many of these abnormalities were attenuated in adult animals, and high doses of leptin partially suppress food intake in the A+P LEPR-KO mice. Although mildly hyperinsulinemic, the A+P LEPR-KO mice displayed normal glucose tolerance and fertility. Thus, AgRP/NPY and POMC neurons each play mandatory roles in aspects of leptin-regulated energy homeostasis, high leptin levels in adult mice mitigate the importance of leptin-responsiveness in these neurons for components of energy balance, suggesting the presence of other leptin-regulated pathways that partially compensate for the lack of leptin action on the POMC and AgRP/NPY neurons.


Assuntos
Proteína Relacionada com Agouti/fisiologia , Ingestão de Alimentos , Metabolismo Energético , Pró-Opiomelanocortina/fisiologia , Receptores para Leptina/fisiologia , Animais , Composição Corporal , Fertilidade , Hiperinsulinismo/etiologia , Hiperfagia , Lactação , Masculino , Camundongos , Neuropeptídeo Y/fisiologia
19.
Cell Metab ; 5(6): 438-49, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17550779

RESUMO

Insulin action in the central nervous system regulates energy homeostasis and glucose metabolism. To define the insulin-responsive neurons that mediate these effects, we generated mice with selective inactivation of the insulin receptor (IR) in either pro-opiomelanocortin (POMC)- or agouti-related peptide (AgRP)-expressing neurons of the arcuate nucleus of the hypothalamus. While neither POMC- nor AgRP-restricted IR knockout mice exhibited altered energy homeostasis, insulin failed to normally suppress hepatic glucose production during euglycemic-hyperinsulinemic clamps in AgRP-IR knockout (IR(DeltaAgRP)) mice. These mice also exhibited reduced insulin-stimulated hepatic interleukin-6 expression and increased hepatic expression of glucose-6-phosphatase. These results directly demonstrate that insulin action in POMC and AgRP cells is not required for steady-state regulation of food intake and body weight. However, insulin action specifically in AgRP-expressing neurons does play a critical role in controlling hepatic glucose production and may provide a target for the treatment of insulin resistance in type 2 diabetes.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Glucose/metabolismo , Insulina/farmacologia , Fígado/metabolismo , Neurônios/efeitos dos fármacos , Animais , Western Blotting , Peso Corporal , Eletrofisiologia , Feminino , Teste de Tolerância a Glucose , Glucose-6-Fosfatase/metabolismo , Homeostase , Hiperinsulinismo/metabolismo , Hipotálamo/metabolismo , Técnicas Imunoenzimáticas , Integrases/metabolismo , Interleucina-6/metabolismo , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptor de Insulina/genética
20.
PLoS Genet ; 3(1): e9, 2007 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-17222061

RESUMO

Genetic variation at the melanocortin 1 receptor (MC1R) is an important risk factor for developing ultraviolet (UV) radiation-induced skin cancer, the most common form of cancer in humans. The underlying mechanisms by which the MC1R defends against UV-induced skin cancer are not known. We used neonatal mouse skin (which, like human skin, contains a mixture of melanocytes and keratinocytes) to study how pigment cells and Mc1r genotype affect the genome-level response to UV radiation. Animals without viable melanocytes (Kit(W-v)/Kit(W-v)) or animals lacking a functional Mc1r (Mc1r(e)/Mc1r(e)) were exposed to sunburn-level doses of UVB radiation, and the patterns of large-scale gene expression in the basal epidermis were compared to each other and to nonmutant animals. Our analysis revealed discrete Kit- and Mc1r-dependent UVB transcriptional responses in the basal epidermis. The Kit-dependent UVB response was characterized largely by an enrichment of oxidative and endoplasmic reticulum stress genes, highlighting a distinctive role for pigmented melanocytes in mediating antioxidant defenses against genotoxic stresses within the basal epidermal environment. By contrast, the Mc1r-dependent UVB response contained an abundance of genes associated with regulating the cell cycle and oncogenesis. To test the clinical relevance of these observations, we analyzed publicly available data sets for primary melanoma and melanoma metastases and found that the set of genes specific for the Mc1r-dependent UVB response was able to differentiate between different clinical subtypes. Our analysis also revealed that the classes of genes induced by UVB differ from those repressed by UVB with regard to their biological functions, their overall number, and their size. The findings described here offer new insights into the transcriptional nature of the UV response in the skin and provide a molecular framework for the underlying mechanisms by which melanocytes and the Mc1r independently mediate and afford protection against UV radiation.


Assuntos
Pigmentação/fisiologia , Pigmentação/efeitos da radiação , Receptor Tipo 1 de Melanocortina/metabolismo , Pele/metabolismo , Pele/efeitos da radiação , Raios Ultravioleta , Animais , Ciclo Celular/efeitos da radiação , Análise por Conglomerados , Epiderme/metabolismo , Epiderme/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Genes Neoplásicos , Genótipo , Humanos , Melanócitos/metabolismo , Melanócitos/efeitos da radiação , Melanoma/genética , Melanoma/patologia , Melanoma/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Pigmentação/genética , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor Tipo 1 de Melanocortina/genética , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos da radiação , Transcrição Gênica/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA