Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Virol ; 93(18)2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31217251

RESUMO

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus. It has a positive-sense RNA genome that also serves as the mRNA for four nonstructural proteins (nsPs) representing subunits of the viral replicase. Coupling of nsP and RNA synthesis complicates analysis of viral RNA replication. We developed trans-replication systems, where production of replication-competent RNA and expression of viral replicase are uncoupled. Mammalian and mosquito RNA polymerase I promoters were used to produce noncapped RNA templates, which are poorly translated relative to CHIKV replicase-generated capped RNAs. It was found that, in human cells, constructs driven by RNA polymerase I promoters of human and Chinese hamster origin performed equally well. In contrast, RNA polymerase I promoters from Aedes mosquitoes exhibited strong species specificity. In both mammalian and mosquito cells, novel trans-replicase assays had exceptional sensitivity, with up to 105-fold higher reporter expression in the presence of replicase relative to background. Using this highly sensitive assay to analyze CHIKV nsP1 functionality, several mutations that severely reduced, but did not completely block, CHIKV replicase activity were identified: (i) nsP1 tagged at its N terminus with enhanced green fluorescent protein; (ii) mutations D63A and Y248A, blocking the RNA capping; and (iii) mutation R252E, affecting nsP1 membrane anchoring. In contrast, a mutation in the nsP1 palmitoylation site completely inactivated CHIKV replicase in both human and mosquito cells and was lethal for the virus. Our data confirm that this novel system provides a valuable tool to study CHIKV replicase, RNA replication, and virus-host interactions.IMPORTANCE Chikungunya virus (CHIKV) is a medically important pathogen responsible for recent large-scale epidemics. The development of efficient therapies against CHIKV has been hampered by gaps in our understanding of how nonstructural proteins (nsPs) function to form the viral replicase and replicate virus RNA. Here we describe an extremely sensitive assay to analyze the effects of mutations on the virus RNA synthesis machinery in cells of both mammalian (host) and mosquito (vector) origin. Using this system, several lethal mutations in CHIKV nsP1 were shown to reduce but not completely block the ability of its replicase to synthesize viral RNAs. However, in contrast to related alphaviruses, CHIKV replicase was completely inactivated by mutations preventing palmitoylation of nsP1. These data can be used to develop novel, virus-specific antiviral treatments.


Assuntos
RNA Polimerase I/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Aedes/virologia , Animais , Antivirais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Febre de Chikungunya/virologia , Vírus Chikungunya/metabolismo , Chlorocebus aethiops , Humanos , Mamíferos/genética , Mosquitos Vetores , Mutação , RNA Polimerase I/fisiologia , RNA Viral/genética , Células Vero , Proteínas não Estruturais Virais/genética
3.
J Cell Sci ; 129(7): 1391-403, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26872785

RESUMO

Integrin-mediated activation of small GTPases induces the polymerisation of G-actin into various actin structures and the release of the transcriptional co-activator MRTF from G-actin. Here we report that pan-integrin-null fibroblasts seeded on fibronectin and expressing ß1- and/or αV-class integrin contained different G-actin pools, nuclear MRTF-A (also known as MKL1 or MAL) levels and MRTF-A-SRF activities. The nuclear MRTF-A levels and activities were highest in cells expressing both integrin classes, lower in cells expressing ß1 integrins and lowest in cells expressing the αV integrins. Quantitative proteomics and transcriptomics analyses linked the differential MRTF-A activities to the expression of the ubiquitin-like modifier interferon-stimulated gene 15 (ISG15), which is known to modify focal adhesion and cytoskeletal proteins. The malignant breast cancer cell line MDA-MB-231 expressed high levels of ß1 integrins, ISG15 and ISGylated proteins, which promoted invasive properties, whereas non-invasive MDA-MB-468 and MCF-7 cell lines expressed low levels of ß1 integrins, ISG15 and ISGylated proteins. Our findings suggest that integrin-adhesion-induced MRTF-A-SRF activation and ISG15 expression constitute a newly discovered signalling circuit that promotes cell migration and invasion.


Assuntos
Neoplasias da Mama/patologia , Citocinas/metabolismo , Integrina alfaV/metabolismo , Integrina beta1/metabolismo , Transativadores/metabolismo , Ubiquitinas/metabolismo , Actinas/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Citocinas/genética , Citoesqueleto/metabolismo , Adesões Focais/metabolismo , Humanos , Células MCF-7 , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Interferência de RNA , RNA Interferente Pequeno/genética , Ubiquitinas/genética
4.
PLoS Genet ; 11(12): e1005710, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26681446

RESUMO

Degeneration of nigrostriatal dopaminergic system is the principal lesion in Parkinson's disease. Because glial cell line-derived neurotrophic factor (GDNF) promotes survival of dopamine neurons in vitro and in vivo, intracranial delivery of GDNF has been attempted for Parkinson's disease treatment but with variable success. For improving GDNF-based therapies, knowledge on physiological role of endogenous GDNF at the sites of its expression is important. However, due to limitations of existing genetic model systems, such knowledge is scarce. Here, we report that prevention of transcription of Gdnf 3'UTR in Gdnf endogenous locus yields GDNF hypermorphic mice with increased, but spatially unchanged GDNF expression, enabling analysis of postnatal GDNF function. We found that increased level of GDNF in the central nervous system increases the number of adult dopamine neurons in the substantia nigra pars compacta and the number of dopaminergic terminals in the dorsal striatum. At the functional level, GDNF levels increased striatal tissue dopamine levels and augmented striatal dopamine release and re-uptake. In a proteasome inhibitor lactacystin-induced model of Parkinson's disease GDNF hypermorphic mice were protected from the reduction in striatal dopamine and failure of dopaminergic system function. Importantly, adverse phenotypic effects associated with spatially unregulated GDNF applications were not observed. Enhanced GDNF levels up-regulated striatal dopamine transporter activity by at least five fold resulting in enhanced susceptibility to 6-OHDA, a toxin transported into dopamine neurons by DAT. Further, we report how GDNF levels regulate kidney development and identify microRNAs miR-9, miR-96, miR-133, and miR-146a as negative regulators of GDNF expression via interaction with Gdnf 3'UTR in vitro. Our results reveal the role of GDNF in nigrostriatal dopamine system postnatal development and adult function, and highlight the importance of correct spatial expression of GDNF. Furthermore, our results suggest that 3'UTR targeting may constitute a useful tool in analyzing gene function.


Assuntos
Dopamina/genética , Neurônios Dopaminérgicos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Doença de Parkinson Secundária/genética , Substância Negra/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/toxicidade , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Humanos , Rim/crescimento & desenvolvimento , Rim/metabolismo , Camundongos , Neostriado/metabolismo , Neostriado/patologia , Fármacos Neuroprotetores/metabolismo , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/patologia , Substância Negra/patologia
5.
Nature ; 461(7266): 1002-6, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19829382

RESUMO

The development of multicellular organisms requires integrin-mediated interactions between cells and their extracellular environment. Integrin binding to extracellular matrix catalyses assembly of multiprotein complexes, which transduce mechanical and chemical signals that regulate many aspects of cell physiology. Integrin-linked kinase (Ilk) is a multifunctional protein that binds beta-integrin cytoplasmic domains and regulates actin dynamics by recruiting actin binding regulatory proteins such as alpha- and beta-parvin. Ilk has also been shown to possess serine/threonine kinase activity and to phosphorylate signalling proteins such as Akt1 and glycogen synthase kinase 3beta (Gsk3beta) in mammalian cells; however, these functions have been shown by genetic studies not to occur in flies and worms. Here we show that mice carrying point mutations in the proposed autophosphorylation site of the putative kinase domain and in the pleckstrin homology domain are normal. In contrast, mice with point mutations in the conserved lysine residue of the potential ATP-binding site of the kinase domain, which mediates Ilk binding to alpha-parvin, die owing to renal agenesis. Similar renal defects occur in alpha-parvin-null mice. Thus, we provide genetic evidence that the kinase activity of Ilk is dispensable for mammalian development; however, an interaction between Ilk and alpha-parvin is critical for kidney development.


Assuntos
Genes Essenciais , Rim/embriologia , Rim/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Proteínas Sanguíneas/química , Movimento Celular , Rim/anormalidades , Lisina/genética , Lisina/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , Mortalidade Perinatal , Fosfoproteínas/química , Fosforilação/genética , Ligação Proteica/genética , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína/genética , Serina/genética , Serina/metabolismo , Análise de Sobrevida
6.
Differentiation ; 81(2): 81-91, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21035938

RESUMO

Semaphorins, originally identified as axon guidance molecules, have also been implicated in angiogenesis, function of the immune system and cancerous growth. Here we show that deletion of Plexin B2 (Plxnb2), a semaphorin receptor that is expressed both in the pretubular aggregates and the ureteric epithelium in the developing kidney, results in renal hypoplasia and occasional double ureters. The rate of cell proliferation in the ureteric epithelium and consequently the number of ureteric tips are reduced in the kidneys lacking Plexin B2 (Plxnb2-/-). Semaphorin 4C, a ligand for Plexin B2, stimulates branching of the ureteric epithelium in wild type and Plxnb2+/- kidney explants, but not in Plxnb2-/- explants. As shown by co-immunoprecipitation Plexin B2 interacts with the Ret receptor tyrosine kinase, the receptor of Glial-cell-line-derived neurotrophic factor (Gdnf), in embryonic kidneys. Isolated Plxnb2-/- ureteric buds fail to respond to Gdnf by branching, but this response is rescued by Fibroblast growth factor 7 and Follistatin as well as by the metanephric mesenchyme. The differentiation of the nephrogenic mesenchyme, its morphology and the rate of apoptosis in the Plxnb2-/- kidneys are normal. Plexin B2 is co-expressed with Plexin B1 (Plxnb1) in the kidney. The double homozygous Plxnb1-Plxnb2-deficient mice show high embryonic lethality prior to onset of nephrogenesis. The only double homozygous embryo surviving to E12 showed hypoplastic kidneys with ureteric branches and differentiating mesenchyme. Taken together, our results show that Sema4C-Plexin B2 signalling regulates ureteric branching, possibly through modulation of Gdnf signalling by interaction with Ret, and suggest non-redundant roles for Plexin B1 and Plexin B2 in kidney development.


Assuntos
Rim/embriologia , Morfogênese/genética , Proteínas do Tecido Nervoso/metabolismo , Semaforinas/metabolismo , Ureter/embriologia , Animais , Diferenciação Celular/genética , Fator 7 de Crescimento de Fibroblastos/genética , Folistatina/farmacologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Rim/anormalidades , Mesoderma/efeitos dos fármacos , Mesoderma/crescimento & desenvolvimento , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Semaforinas/genética , Ureter/anormalidades , Urotélio/efeitos dos fármacos , Urotélio/embriologia
7.
J Am Soc Nephrol ; 22(4): 718-31, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21436291

RESUMO

Renal dysplasia, defined by defective ureteric branching morphogenesis and nephrogenesis, is the major cause of renal failure in infants and children. Here, we define a pathogenic role for a ß-catenin-activated genetic pathway in murine renal dysplasia. Stabilization of ß-catenin in the ureteric cell lineage before the onset of kidney development increased ß-catenin levels and caused renal aplasia or severe hypodysplasia. Analysis of gene expression in the dysplastic tissue identified downregulation of genes required for ureteric branching and upregulation of Tgfß2 and Dkk1. Treatment of wild-type kidney explants with TGFß2 or DKK1 generated morphogenetic phenotypes strikingly similar to those observed in mutant kidney tissue. Stabilization of ß-catenin after the onset of kidney development also caused dysplasia and upregulation of Tgfß2 and Dkk1 in the epithelium. Together, these results demonstrate that elevation of ß-catenin levels during kidney development causes dysplasia.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Rim/anormalidades , Rim/embriologia , Fator de Crescimento Transformador beta2/fisiologia , Regulação para Cima/fisiologia , beta Catenina/fisiologia , Animais , Apoptose/fisiologia , Proliferação de Células , Modelos Animais de Doenças , Feminino , Rim/fisiopatologia , Camundongos , Camundongos Mutantes , Morfogênese/fisiologia , Gravidez , Transdução de Sinais/fisiologia , Ureter/anormalidades , Ureter/embriologia , Ureter/fisiopatologia , Proteínas Wnt/fisiologia
8.
J Am Soc Nephrol ; 22(2): 274-84, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21289216

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) is indispensable for ureteric budding and branching. If applied exogenously, GDNF promotes ectopic ureteric buds from the Wolffian duct. Although several downstream effectors of GDNF are known, the identification of early response genes is incomplete. Here, microarray screening detected several GDNF-regulated genes in the Wolffian duct, including Visinin like 1 (Vsnl1), which encodes a neuronal calcium-sensor protein. We observed renal Vsnl1 expression exclusively in the ureteric epithelium, but not in Gdnf-null kidneys. In the tissue culture of Gdnf-deficient kidney primordium, exogenous GDNF and alternative bud inducers (FGF7 and follistatin) restored Vsnl1 expression. Hence, Vsnl1 characterizes the tip of the ureteric bud epithelium regardless of the inducer. In the tips, Vsnl1 showed a mosaic expression pattern that was mutually exclusive with ß-catenin transcriptional activation. Vsnl1 was downregulated in both ß-catenin-stabilized and ß-catenin-deficient kidneys. Moreover, in a mouse collecting duct cell line, Vsnl1 compromised ß-catenin stability, suggesting a counteracting relationship between Vsnl1 and ß-catenin. In summary, Vsnl1 marks ureteric bud tips in embryonic kidneys, and its mosaic pattern demonstrates a heterogeneity of cell types that may be critical for normal ureteric branching.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Neurocalcina/fisiologia , Ureter/embriologia , Animais , Biomarcadores , Cálcio/metabolismo , Ciclo Celular , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , beta Catenina/fisiologia
9.
Sci Rep ; 9(1): 5302, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30923332

RESUMO

Mechanisms controlling ureter lenght and the position of the kidney are poorly understood. Glial cell-line derived neurotrophic factor (GDNF) induced RET signaling is critical for ureteric bud outgrowth, but the function of endogenous GDNF in further renal differentiation and urogenital system development remains discursive. Here we analyzed mice where 3' untranslated region (UTR) of GDNF is replaced with sequence less responsive to microRNA-mediated regulation, leading to increased GDNF expression specifically in cells naturally transcribing Gdnf. We demonstrate that increased Gdnf leads to short ureters in kidneys located in an abnormally caudal position thus resembling human pelvic kidneys. High GDNF levels expand collecting ductal progenitors at the expense of ureteric trunk elongation and result in expanded tip and short trunk phenotype due to changes in cell cycle length and progenitor motility. MEK-inhibition rescues these defects suggesting that MAPK-activity mediates GDNF's effects on progenitors. Moreover, Gdnf   hyper mice are infertile likely due to effects of excess GDNF on distal ureter remodeling. Our findings suggest that dysregulation of GDNF levels, for example via alterations in 3'UTR, may account for a subset of congenital anomalies of the kidney and urinary tract (CAKUT) and/or congenital infertility cases in humans and pave way to future studies.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Infertilidade/genética , Anormalidades Urogenitais/genética , Refluxo Vesicoureteral/genética , Regiões 3' não Traduzidas/genética , Animais , Apoptose/genética , Ciclo Celular/genética , Movimento Celular/genética , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Infertilidade/congênito , Infertilidade/patologia , Rim/anormalidades , Rim/embriologia , Rim/patologia , Masculino , Camundongos , Camundongos Transgênicos , MicroRNAs/metabolismo , Técnicas de Cultura de Órgãos , Transdução de Sinais/genética , Células-Tronco/fisiologia , Ureter/anormalidades , Ureter/embriologia , Ureter/patologia , Anormalidades Urogenitais/patologia , Refluxo Vesicoureteral/patologia
10.
Nat Med ; 20(4): 350-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24681597

RESUMO

Kindlin-1 is an integrin tail binding protein that controls integrin activation. Mutations in the FERMT-1 gene, which encodes for Kindlin-1, lead to Kindler syndrome in man, which is characterized by skin blistering, premature skin aging and skin cancer of unknown etiology. Here we show that loss of Kindlin-1 in mouse keratinocytes recapitulates Kindler syndrome and also produces enlarged and hyperactive stem cell compartments, which lead to hyperthickened epidermis, ectopic hair follicle development and increased skin tumor susceptibility. Mechanistically, Kindlin-1 controls keratinocyte adhesion through ß1-class integrins and proliferation and differentiation of cutaneous epithelial stem cells by promoting α(v)ß(6) integrin-mediated transforming growth factor-ß (TGF-ß) activation and inhibiting Wnt-ß-catenin signaling through integrin-independent regulation of Wnt ligand expression. Our findings assign Kindlin-1 the previously unknown and essential task of controlling cutaneous epithelial stem cell homeostasis by balancing TGF-ß-mediated growth-inhibitory signals and Wnt-ß-catenin-mediated growth-promoting signals.


Assuntos
Vesícula , Proteínas de Transporte/fisiologia , Proliferação de Células , Epidermólise Bolhosa , Queratinócitos/metabolismo , Doenças Periodontais , Transtornos de Fotossensibilidade , Pele/citologia , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Proteínas de Transporte/genética , Adesão Celular/genética , Adesão Celular/fisiologia , Modelos Animais de Doenças , Folículo Piloso/patologia , Integrina beta1/metabolismo , Integrinas/metabolismo , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Pele/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
11.
J Am Soc Nephrol ; 18(4): 1130-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17329570

RESUMO

Wnt proteins are required for induction of nephrons in mouse metanephric kidneys, but the downstream pathways that mediate tubule induction and epithelial differentiation have remained obscure. The intracellular mechanisms by which Wnt signaling mediates nephron induction in embryonic kidney mesenchymes were studied. First is shown that transient exposure of isolated kidney mesenchymes to structurally different glycogen synthase kinase-3 (GSK3) inhibitors lithium or 6-bromoindirubin-3'-oxime results in abundant epithelial differentiation and full segregation of nephrons. Shown further by mice with genetically disrupted ureteric bud or Wolffian duct development is that this nephrogenic competence arises independent of the influence of Wolffian duct-derived epithelia. Analysis of the intracellular signaling cascades downstream of GSK3 inhibition revealed stabilization of beta-catenin and upregulation of Lef1 and Tcf1, both events that are associated with the active canonical Wnt signaling. Last, genetic evidence that metanephric mesenchyme-specific stabilization of beta-catenin is sufficient to induce nephron differentiation in isolated kidney mesenchymes, similar to that induced by GSK3 inhibitors, is provided. These data show that activation of canonical Wnt pathway is sufficient to induce nephrogenesis and suggest that this pathway mediates the nephron induction in murine kidney mesenchymes.


Assuntos
Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Rim/embriologia , Mesoderma/citologia , Néfrons/embriologia , beta Catenina/metabolismo , Animais , Diferenciação Celular , Fator 1 de Ligação ao Facilitador Linfoide/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Wistar , Proteínas Wnt/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa