Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Invest Dermatol ; 140(3): 615-623.e5, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31479664

RESUMO

Abundant corneocyte surface protrusions, observed in patients with atopic dermatitis with filaggrin loss-of-function mutations, are inversely associated with levels of natural moisturizing factors (NMFs) in the stratum corneum. To dissect the etiological role of NMFs and filaggrin deficiency in surface texture alterations, we examined mouse models with genetic deficiencies in the synthesis or degradation of filaggrin monomers for NMFs, cell stiffness (elastic modulus) and corneocyte surface protrusion density (dermal texture index). Five neonatal and adult mouse models carrying inactivating mutations of SASPase (Sasp-/-), filaggrin (Flgft/ft and Flg-/-), filaggrin-hornerin (FlgHrnr-/-), and bleomycin hydrolase (Blmh-/-) were investigated. Sasp-/- and Flg-/- were on the hairless mouse background. Atomic force microscopy was used to determine elastic modulus and dermal texture index. Corneocytes of each neonatal as well as hairless adult knockout mouse exhibited an increased number of protrusions and decreased elastic modulus. In these mice, NMFs were reduced except for Sasp-/-. Dermal texture index was inversely correlated with NMFs and elastic modulus. Our findings demonstrate that any filaggrin-NMF axis deficiency can affect corneocyte mechanical properties in mice and likely in humans. Differences in NMFs and corneocyte surface texture between neonatal and adult as well as hairless and hairy mice emphasize the need for carefully selecting the most appropriate animal models for studies.


Assuntos
Dermatite Atópica/patologia , Células Epidérmicas/patologia , Epiderme/patologia , Proteínas de Filamentos Intermediários/deficiência , Animais , Ácido Aspártico Endopeptidases/genética , Cisteína Endopeptidases/genética , Dermatite Atópica/genética , Modelos Animais de Doenças , Módulo de Elasticidade , Células Epidérmicas/ultraestrutura , Proteínas Filagrinas , Humanos , Proteínas de Filamentos Intermediários/genética , Mutação com Perda de Função , Camundongos , Camundongos Knockout , Microscopia de Força Atômica
2.
Front Immunol ; 9: 988, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867965

RESUMO

Mutations in the filaggrin gene (Flg) are associated with increased systemic levels of Th17 cells and increased IL-17A production following antigen exposure in both humans and mice. In addition to Th17 cells, γδ T cells can produce IL-17A. The differentiation of γδ T cells to either IFNγ or IL-17A-producing (γδT17) cells is mainly determined in the thymus. Interestingly, it has been reported that filaggrin is expressed in the Hassall bodies in the human thymic medulla. However, whether filaggrin affects γδ T cell development is not known. Here, we show that filaggrin-deficient flaky tail (ft/ft) mice have an increased number of γδT17 cells in the spleen, epidermis, and thymus compared to wild-type (WT) mice. We demonstrate that filaggrin is expressed in the mouse thymic medulla and that blocking the egress of cells from the thymus results in accumulation of Vγ2+ γδT17 cells in the thymus of adult ft/ft mice. Finally, we find increased T cell receptor expression levels on γδ T cells and increased levels of IL-6 and IL-23 in the thymus of ft/ft mice. These findings demonstrate that filaggrin is expressed in the mouse thymic medulla and that production of Vγ2+ γδT17 cells is dysregulated in filaggrin-deficient ft/ft mice.


Assuntos
Interleucina-17/imunologia , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Células Th17/imunologia , Timo/citologia , Animais , Diferenciação Celular , Proteínas Filagrinas , Interleucina-23/genética , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Pele/imunologia , Baço/imunologia , Timo/imunologia
3.
J Immunol ; 199(2): 707-717, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28615416

RESUMO

Atopic dermatitis (AD) is a common inflammatory skin disease affecting up to 20% of children and 3% of adults worldwide and is associated with dysregulation of the skin barrier. Although type 2 responses are implicated in AD, emerging evidence indicates a potential role for the IL-17A signaling axis in AD pathogenesis. In this study we show that in the filaggrin mutant mouse model of spontaneous AD, IL-17RA deficiency (Il17ra-/- ) resulted in severe exacerbation of skin inflammation. Interestingly, Il17ra-/- mice without the filaggrin mutation also developed spontaneous progressive skin inflammation with eosinophilia, as well as increased levels of thymic stromal lymphopoietin (TSLP) and IL-5 in the skin. Il17ra-/- mice have a defective skin barrier with altered filaggrin expression. The barrier dysregulation and spontaneous skin inflammation in Il17ra-/- mice was dependent on TSLP, but not the other alarmins IL-25 and IL-33. The associated skin inflammation was mediated by IL-5-expressing pathogenic effector Th2 cells and was independent of TCRγδ T cells and IL-22. An absence of IL-17RA in nonhematopoietic cells, but not in the hematopoietic cells, was required for the development of spontaneous skin inflammation. Skin microbiome dysbiosis developed in the absence of IL-17RA, with antibiotic intervention resulting in significant amelioration of skin inflammation and reductions in skin-infiltrating pathogenic effector Th2 cells and TSLP. This study describes a previously unappreciated protective role for IL-17RA signaling in regulation of the skin barrier and maintenance of skin immune homeostasis.


Assuntos
Dermatite Atópica/imunologia , Receptores de Interleucina-17/imunologia , Receptores de Interleucina-17/metabolismo , Pele/crescimento & desenvolvimento , Pele/patologia , Animais , Citocinas/imunologia , Dermatite Atópica/patologia , Modelos Animais de Doenças , Disbiose , Eosinofilia/imunologia , Proteínas Filagrinas , Regulação da Expressão Gênica , Homeostase , Interleucina-33/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Interleucinas/genética , Interleucinas/imunologia , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/genética , Camundongos , Microbiota , Mutação , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Interleucina-17/deficiência , Receptores de Interleucina-17/genética , Transdução de Sinais , Pele/imunologia , Pele/microbiologia , Células Th2/imunologia , Linfopoietina do Estroma do Timo , Interleucina 22
4.
Clin Exp Dermatol ; 42(6): 622-631, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28556377

RESUMO

BACKGROUND: Atopic dermatitis (AD) is a common inflammatory skin disorder, characterized by skin barrier defects and enhanced allergen priming. Null mutations in the filaggrin gene (FLG) are strongly associated with moderate to severe AD, but the pathways linking barrier dysfunction and cutaneous inflammation are still largely unknown. AIM: To assess alteration of endogenous cysteine protease activity in FLG-deficient keratinocytes, and to determine whether the alteration in cysteine protease activity affects epidermal barrier function and associated gene and protein expression. METHODS: We established a stable FLG knockdown cell line, and reconstructed epidermal equivalents in vitro. Barrier function of the reconstructed epidermis, the barrier-associated genes and proteins, and the activity of endogenous cysteine proteases were tested. Inhibitors of cysteine proteases were used to further evaluate the role of endogenous cysteine proteases in epidermal barrier function. RESULTS: FLG knockdown induced impaired epidermal barrier function. Microarray, western blotting and fluorescence staining showed reduced expression of K10, ZO-1, E-cadherin, claudin-1 and occludin in FLG knockdown keratinocytes. Compared with cysteine protease activity in control cells, protease activity was dramatically enhanced in FLG knockdown keratinocytes. Furthermore, administration of cysteine protease inhibitors significantly recovered expression of K10 and tight junction proteins, and the barrier defect induced by FLG deficiency. CONCLUSIONS: This is the first observation of elevated endogenous cysteine protease activity in FLG-deficient keratinocytes, which may play an important role in impaired barrier function in AD skin. Modulation of cysteine protease activity might be a novel therapeutic approach for AD treatment.


Assuntos
Cisteína Proteases/metabolismo , Dermatite Atópica/metabolismo , Proteínas de Filamentos Intermediários/deficiência , Queratinócitos/metabolismo , Absorção Cutânea/fisiologia , Linhagem Celular , Proliferação de Células , Dermatite Atópica/fisiopatologia , Proteínas Filagrinas , Humanos , Queratinócitos/citologia , Queratinas/metabolismo
5.
Am J Pathol ; 185(4): 1012-21, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25660180

RESUMO

Although keratosis pilaris (KP) is common, its etiopathogenesis remains unknown. KP is associated clinically with ichthyosis vulgaris and atopic dermatitis and molecular genetically with filaggrin-null mutations. In 20 KP patients and 20 matched controls, we assessed the filaggrin and claudin 1 genotypes, the phenotypes by dermatoscopy, and the morphology by light and transmission electron microscopy. Thirty-five percent of KP patients displayed filaggrin mutations, demonstrating that filaggrin mutations only partially account for the KP phenotype. Major histologic and dermatoscopic findings of KP were hyperkeratosis, hypergranulosis, mild T helper cell type 1-dominant lymphocytic inflammation, plugging of follicular orifices, striking absence of sebaceous glands, and hair shaft abnormalities in KP lesions but not in unaffected skin sites. Changes in barrier function and abnormal paracellular permeability were found in both interfollicular and follicular stratum corneum of lesional KP, which correlated ultrastructurally with impaired extracellular lamellar bilayer maturation and organization. All these features were independent of filaggrin genotype. Moreover, ultrastructure of corneodesmosomes and tight junctions appeared normal, immunohistochemistry for claudin 1 showed no reduction in protein amounts, and molecular analysis of claudin 1 was unremarkable. Our findings suggest that absence of sebaceous glands is an early step in KP pathogenesis, resulting in downstream hair shaft and epithelial barrier abnormalities.


Assuntos
Anormalidades Múltiplas/patologia , Doença de Darier/patologia , Epiderme/anormalidades , Sobrancelhas/anormalidades , Cabelo/anormalidades , Proteínas de Filamentos Intermediários/deficiência , Glândulas Sebáceas/anormalidades , Anormalidades Múltiplas/genética , Adulto , Idoso , Claudina-1/metabolismo , Doença de Darier/genética , Dermoscopia , Desmossomos/metabolismo , Epiderme/ultraestrutura , Sobrancelhas/patologia , Feminino , Proteínas Filagrinas , Genótipo , Cabelo/ultraestrutura , Humanos , Proteínas de Filamentos Intermediários/genética , Masculino , Pessoa de Meia-Idade , Mutação/genética , Permeabilidade , Fenótipo , Glândulas Sebáceas/patologia , Glândulas Sebáceas/ultraestrutura , Adulto Jovem
6.
Br J Dermatol ; 171(4): 771-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24958284

RESUMO

BACKGROUND: Histamine is an abundant mediator accumulating in the skin of atopic patients, where it is thought to be derived from immune cells. While keratinocytes express histidine decarboxylase (HDC), levels of the enzyme in normal or diseased epidermis and factors that influence its expression in human keratinocytes are not known. OBJECTIVES: To assess levels of HDC in inflammatory skin diseases and factors influencing its expression. METHODS: Normal and filaggrin-insufficient human keratinocytes, organotypic epidermal models and skin samples were investigated for the expression of HDC. The effect of cytokines, bacterial and allergen stimuli exposure and functional changes in differentiation were evaluated in vitro. RESULTS: We detected abundant expression of the HDC protein in all models studied; expression was increased in atopic skin samples. Filaggrin-insufficient keratinocytes maintained HDC levels, but exposure of keratinocytes to thymic stromal lymphopoietin, tumour necrosis factor-α, lipopolysaccharide (LPS) and house dust mite (HDM) extract increased HDC expression in vitro. Furthermore, filaggrin expression in cultured keratinocytes increased following histamine depletion. CONCLUSIONS: Keratinocytes express abundant HDC protein, and the levels increase in atopic skin. LPS, HDM and cytokines, which are implicated in allergic inflammation, promote the expression of the enzyme and upregulate histamine levels in keratinocytes. Actively produced histamine influences keratinocyte differentiation, suggesting functional relevance of the axis to atopic dermatitis. The findings therefore identify a new point of therapeutic intervention.


Assuntos
Dermatite Atópica/enzimologia , Histidina Descarboxilase/metabolismo , Queratinócitos/enzimologia , Diferenciação Celular/fisiologia , Células Cultivadas , Citocinas/fisiologia , Eczema/enzimologia , Epiderme/enzimologia , Proteínas Filagrinas , Humanos , Proteínas de Filamentos Intermediários/deficiência , Regulação para Cima
8.
Nature ; 495(7440): 227-30, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23434756

RESUMO

Haematopoietic stem cells (HSCs) primarily reside in the bone marrow where signals generated by stromal cells regulate their self-renewal, proliferation and trafficking. Endosteal osteoblasts and perivascular stromal cells including endothelial cells, CXCL12-abundant reticular cells, leptin-receptor-positive stromal cells, and nestin-green fluorescent protein (GFP)-positive mesenchymal progenitors have all been implicated in HSC maintenance. However, it is unclear whether specific haematopoietic progenitor cell (HPC) subsets reside in distinct niches defined by the surrounding stromal cells and the regulatory molecules they produce. CXCL12 (chemokine (C-X-C motif) ligand 12) regulates both HSCs and lymphoid progenitors and is expressed by all of these stromal cell populations. Here we selectively deleted Cxcl12 from candidate niche stromal cell populations and characterized the effect on HPCs. Deletion of Cxcl12 from mineralizing osteoblasts has no effect on HSCs or lymphoid progenitors. Deletion of Cxcl12 from osterix-expressing stromal cells, which include CXCL12-abundant reticular cells and osteoblasts, results in constitutive HPC mobilization and a loss of B-lymphoid progenitors, but HSC function is normal. Cxcl12 deletion from endothelial cells results in a modest loss of long-term repopulating activity. Strikingly, deletion of Cxcl12 from nestin-negative mesenchymal progenitors using Prx1-cre (Prx1 also known as Prrx1) is associated with a marked loss of HSCs, long-term repopulating activity, HSC quiescence and common lymphoid progenitors. These data suggest that osterix-expressing stromal cells comprise a distinct niche that supports B-lymphoid progenitors and retains HPCs in the bone marrow, and that expression of CXCL12 from stromal cells in the perivascular region, including endothelial cells and mesenchymal progenitors, supports HSCs.


Assuntos
Quimiocina CXCL2/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Linfócitos B/citologia , Medula Óssea/metabolismo , Movimento Celular , Quimiocina CXCL2/deficiência , Quimiocina CXCL2/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Filamentos Intermediários/deficiência , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Proteínas do Tecido Nervoso/deficiência , Nestina , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Nicho de Células-Tronco/fisiologia
9.
J Neuropathol Exp Neurol ; 71(12): 1047-63, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23147502

RESUMO

Interactions between the embryonic pial basement membrane (PBM) and radial glia (RG) are essential for morphogenesis of the cerebral cortex because disrupted interactions cause cobblestone malformations. To elucidate the role of dystroglycan (DG) in PBM-RG interactions, we studied the expression of DG protein and Dag1 mRNA (which encodes DG protein) in developing cerebral cortex and analyzed cortical phenotypes in Dag1 CNS conditional mutant mice. In normal embryonic cortex, Dag1 mRNA was expressed in the ventricular zone, which contains RG nuclei, whereas DG protein was expressed at the cortical surface on RG end feet. Breaches of PBM continuity appeared during early neurogenesis in Dag1 mutants. Diverse cellular elements streamed through the breaches to form leptomeningeal heterotopia that were confluent with the underlying residual cortical plate and contained variably truncated RG fibers, many types of cortical neurons, and radial and intermediate progenitor cells. Nevertheless, layer-specific molecular expression seemed normal in heterotopic neurons, and axons projected to appropriate targets. Dendrites, however, were excessively tortuous and lacked radial orientation. These findings indicate that DG is required on RG end feet to maintain PBM integrity and suggest that cobblestone malformations involve disturbances of RG structure, progenitor distribution, and dendrite orientation, in addition to neuronal "overmigration."


Assuntos
Membrana Basal , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Distroglicanas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Neuroglia/citologia , Fatores Etários , Animais , Membrana Basal/citologia , Membrana Basal/embriologia , Membrana Basal/metabolismo , Bromodesoxiuridina/metabolismo , Movimento Celular/genética , Proliferação de Células , Distroglicanas/genética , Embrião de Mamíferos , Feminino , Marcação In Situ das Extremidades Cortadas , Proteínas de Filamentos Intermediários/deficiência , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/fisiologia , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Células-Tronco/fisiologia , Proteínas Supressoras de Tumor/metabolismo
10.
Am J Pathol ; 181(3): 969-77, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22796440

RESUMO

Disruption of skin barrier function leads to increases in the percutaneous transfer of allergens and the incidence of atopic dermatitis. Flaky tail (Flg(ft)) mice have been used as a model of atopic dermatitis with skin barrier dysfunction. Although Flg(ft) mice are known to have filaggrin mutation, the mechanism responsible for the skin barrier dysfunction that they display needs to be determined, especially for the roles of epidermal adhesion and junction proteins. Herein, we report the decreased expression of epidermal growth factor receptor (EGFR), E-cadherin, occludin, and SIRT1 in the skin of Flg(ft) mice, compared with those in C57BL/6J mice. Administration of N-acetyl-L-cysteine, an antioxidant, in the drinking water improved these protein expressions in the skin of Flg(ft) mice. Notably, we discovered that loricrin expression was suppressed in Flg(ft) mice. In vitro experiments showed that filaggrin small interfering RNA, loricrin small interfering RNA, or SIRT1 inhibitor sirtinol suppressed the expression levels of EGFR, E-cadherin, and occludin in a human immortalized keratinocyte cell line (HaCaT cells). Our findings suggest that the observed reductions in EGFR, E-cadherin, and occludin expression were due to filaggrin deficiency accompanied with subsequent loricrin deficiency and disruption of the SIRT1 pathway in the skin of Flg(ft) mice.


Assuntos
Caderinas/metabolismo , Receptores ErbB/metabolismo , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Pele/metabolismo , Cauda/patologia , Acetilcisteína/farmacologia , Ar , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cruzamentos Genéticos , Citocinas/metabolismo , Feminino , Proteínas Filagrinas , Humanos , Mediadores da Inflamação/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ocludina , RNA Interferente Pequeno/metabolismo , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo , Pele/efeitos dos fármacos , Pele/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
J Neuroinflammation ; 9: 61, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22458306

RESUMO

BACKGROUND: Mice with peroxisome deficiency in neural cells (Nestin-Pex5-/-) develop a neurodegenerative phenotype leading to motor and cognitive disabilities and early death. Major pathologies at the end stage of disease include severe demyelination, axonal degeneration and neuroinflammation. We now investigated the onset and progression of these pathological processes, and their potential interrelationship. In addition, the putative role of oxidative stress, the impact of plasmalogen depletion on the neurodegenerative phenotype, and the consequences of peroxisome elimination in the postnatal period were studied. METHODS: Immunohistochemistry in association with gene expression analysis was performed on Nestin-Pex5-/- mice to document demyelination, axonal damage and neuroinflammation. Also Gnpat-/- mice, with selective plasmalogen deficiency and CMV-Tx-Pex5-/- mice, with tamoxifen induced generalized loss of peroxisomes were analysed. RESULTS: Activation of the innate immune system is a very early event in the pathological process in Nestin-Pex5-/- mice which evolves in chronic neuroinflammation. The complement factor C1q, one of the earliest up regulated transcripts, was expressed on neurons and oligodendrocytes but not on microglia. Transcripts of other pro- and anti-inflammatory genes and markers of phagocytotic activity were already significantly induced before detecting pathologies with immunofluorescent staining. Demyelination, macrophage activity and axonal loss co-occurred throughout the brain. As in patients with mild peroxisome biogenesis disorders who develop regressive changes, demyelination in cerebellum and brain stem preceded major myelin loss in corpus callosum of both Nestin-Pex5-/- and CMV-Tx-Pex5-/- mice. These lesions were not accompanied by generalized oxidative stress throughout the brain. Although Gnpat-/- mice displayed dysmyelination and Purkinje cell axon damage in cerebellum, confirming previous observations, no signs of inflammation or demyelination aggravating with age were observed. CONCLUSIONS: Peroxisome inactivity triggers a fast neuroinflammatory reaction, which is not solely due to the depletion of plasmalogens. In association with myelin abnormalities this causes axon damage and loss.


Assuntos
Axônios/fisiologia , Sistema Nervoso Central/fisiologia , Imunidade Inata/fisiologia , Plasmalogênios/biossíntese , Receptores Citoplasmáticos e Nucleares/deficiência , Animais , Axônios/patologia , Sistema Nervoso Central/patologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Proteínas de Filamentos Intermediários/deficiência , Metabolismo dos Lipídeos/fisiologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Nestina , Receptor 1 de Sinal de Orientação para Peroxissomos , Peroxissomos/metabolismo
12.
Brain ; 134(Pt 7): 1943-58, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21705419

RESUMO

The polyglutamine diseases consist of nine neurodegenerative disorders including spinocerebellar ataxia type 17 that is caused by a polyglutamine tract expansion in the TATA box-binding protein. In all polyglutamine diseases, polyglutamine-expanded proteins are ubiquitously expressed throughout the body but cause selective neurodegeneration. Understanding the specific effects of polyglutamine-expanded proteins, when expressed at the endogenous levels, in neurons is important for unravelling the pathogenesis of polyglutamine diseases. However, addressing this important issue using mouse models that either overly or ubiquitously express mutant polyglutamine proteins in the brain and body has proved difficult. To investigate the pathogenesis of spinocerebellar ataxia 17, we generated a conditional knock-in mouse model that expresses one copy of the mutant TATA box-binding protein gene, which encodes a 105-glutamine repeat, selectively in neuronal cells at the endogenous level. Neuronal expression of mutant TATA box-binding protein causes age-dependent neurological symptoms in mice and the degeneration of cerebellar Purkinje cells. Mutant TATA box-binding protein binds more tightly to the transcription factor nuclear factor-Y, inhibits its association with the chaperone protein promoter, as well as the promoter activity and reduces the expression of the chaperones Hsp70, Hsp25 and HspA5, and their response to stress. These findings demonstrate how mutant TATA box-binding protein at the endogenous level affects neuronal function, with important implications for the pathogenesis and treatment of polyglutamine diseases.


Assuntos
Encéfalo/patologia , Fator de Ligação a CCAAT/metabolismo , Chaperonas Moleculares/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Peptídeos/metabolismo , Proteína de Ligação a TATA-Box/metabolismo , Fatores Etários , Animais , Peso Corporal/genética , Encéfalo/metabolismo , Fator de Ligação a CCAAT/genética , Linhagem Celular Transformada , Imunoprecipitação da Cromatina/métodos , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Feminino , Regulação da Expressão Gênica/genética , Humanos , Proteínas de Filamentos Intermediários/deficiência , Masculino , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Proteínas do Tecido Nervoso/deficiência , Nestina , Doenças Neurodegenerativas/genética , Neurônios/patologia , Peptídeos/genética , Proteína de Ligação a TATA-Box/genética , Transfecção
13.
Am J Physiol Cell Physiol ; 294(5): C1183-91, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18353892

RESUMO

The inflammatory response is thought to play important roles in tissue healing. The hypothesis of this study was that the inflammatory cytokine interferon (IFN)-gamma is produced endogenously following skeletal muscle injury and promotes efficient healing. We show that IFN-gamma is expressed at both mRNA and protein levels in skeletal muscle following injury, and that the time course of IFN-gamma expression correlated with the accumulation of macrophages, T-cells, and natural killer cells, as well as myoblasts, in damaged muscle. Cells of each type were isolated from injured muscle, and IFN-gamma expression was detected in each cell type. We also demonstrate that administration of an IFN-gamma receptor blocking antibody to wild-type mice impaired induction of interferon response factor-1, reduced cell proliferation, and decreased formation of regenerating fibers. IFN-gamma null mice showed similarly impaired muscle healing associated with impaired macrophage function and development of fibrosis. In vitro studies demonstrated that IFN-gamma and its receptor are expressed in the C2C12 muscle cell line, and that the IFN-gamma receptor blocking antibody reduced proliferation and fusion of these muscle cells. In summary, our results indicate that IFN-gamma promotes muscle healing, in part, by stimulating formation of new muscle fibers.


Assuntos
Interferon gama/fisiologia , Músculo Esquelético/fisiologia , Mioblastos/fisiologia , Regeneração , Animais , Técnicas de Cultura de Células , Divisão Celular , Proteínas Cardiotóxicas de Elapídeos/toxicidade , Primers do DNA , Membro Anterior , Inflamação , Interferon gama/deficiência , Interferon gama/genética , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Musculares Esqueléticas/fisiologia , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Receptores de Interferon/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor de Interferon gama
14.
Stem Cells ; 26(4): 1027-36, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18276799

RESUMO

Sonic hedgehog (Shh) is necessary for sustaining the proliferation of neural stem cells (NSCs), yet little is known about its mechanisms. Whereas Gli1, Gli2, and Gli3, the primary mediators of Shh signaling, were all expressed in hippocampal neural progenitors, Shh treatment of NSCs induced only Gli1 expression. Acute depletion of Gli1 in postnatal NSCs by short-hairpin RNA decreased proliferation, whereas germline deletion of Gli1 did not affect NSC proliferation, suggesting a difference in mechanisms of Gli1 compensation that may be developmentally dependent. To determine whether Gli1 was sufficient to enhance NSC proliferation, we overexpressed this mitogen and were surprised to find that Gli1 resulted in decreased proliferation, accumulation of NSCs in the G2/M phase of cell cycle, and apoptosis. In contrast, Gli1-expressing lineage-restricted neural precursors demonstrated a 4.5-fold proliferation enhancement. Expression analyses of Gli1-expressing NSCs identified significant induction of Gadd45a and decreased cyclin A2 and Stag1 mRNA, genes involved in the G2-M transition and apoptosis. Furthermore, Gadd45a overexpression was sufficient to partially recapitulate the Gli1-induced G2/M accumulation and cell death of NSCs. In contrast to normal stem cells, tumor-derived stem cells had markedly higher basal Gli1 expression and did not undergo apoptosis with further elevation of Gli1. Our data suggest that Gli1-induced apoptosis may serve as a protective mechanism against premature mitosis and may give insight into mechanisms by which nonmalignant stem cells restrain hyperproliferation in the context of potentially transforming mitogenic signals. Tumor-derived stem cells apparently lack these mechanisms, which may contribute to their unrestrained proliferation and malignant potential.


Assuntos
Apoptose/fisiologia , Divisão Celular/fisiologia , Fase G2/fisiologia , Hipocampo/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Células-Tronco Neoplásicas/patologia , Neurônios/patologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Divisão Celular/genética , Células Cultivadas , Fase G2/genética , Hipocampo/metabolismo , Hipocampo/patologia , Proteínas de Filamentos Intermediários/biossíntese , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/genética , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Nestina , Neurônios/citologia , Neurônios/metabolismo , Células Tumorais Cultivadas , Proteína GLI1 em Dedos de Zinco
15.
Dermatitis ; 19(6): 303-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19134432

RESUMO

Atopic dermatitis (AD) is a chronic relapsing inflammatory skin disease. Recently, it was demonstrated that a large proportion of individuals with AD have an epidermal expression deficiency of filaggrin (FLG). This unique finding may have great implications for our understanding of nickel sensitization because nickel is chelated in the epidermis and perhaps to FLG. This review aims to briefly present the current knowledge about nickel sensitization in relation to FLG deficiency and speculate on its possible implications. The new knowledge concerning loss-of-function mutations in the FLG gene (the lack of specific nickel-chelating power in the stratum corneum and a generally defective skin barrier) suggests that an additive effect from irritants and nickel may aggravate hand eczema in individuals with loss-of-function mutations in the FLG gene. This hypothesis calls for a reevaluation of the potential risk of nickel sensitization through the establishment of cohorts with and without such mutations.


Assuntos
Dermatite Atópica/etiologia , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/genética , Metais/efeitos adversos , Níquel/efeitos adversos , Dermatite Atópica/genética , Proteínas Filagrinas , Mãos , Humanos , Mutação
16.
Mol Cell Neurosci ; 25(1): 103-10, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14962744

RESUMO

Inherited retinal degenerations are a major cause of blindness for which there are currently no effective therapies. Significant progress concerning in vivo gene transfer has allowed retardation of degeneration or retinal functional improvement in different animal models. To date, there has been no evaluation of the impact of these treatments on higher visual function, a critical step for validating gene therapy treatment strategies. Here, we have used adeno-associated (AAV2)-mediated gene transfer of Prph2 in the Prph2(Rd2/Rd2) mouse model. We then assessed higher visual function by recording from central visually responsive neurons in the superior colliculus and improvements were correlated in individual animals with retinal function (ERG) and histological and biochemical changes. Although gene replacement therapy only partially restores photoreceptor morphology, it results in a 300% increase of the visual cycle protein rhodopsin, leading to retinal function improvement (250% increase of b-wave amplitude) and significantly higher central visual responses (166% increase at 24 cd/m(2)). These findings suggest that gene replacement therapy leading to even relatively modest structural improvement may result in improved central visual function.


Assuntos
Terapia Genética/métodos , Proteínas de Filamentos Intermediários/deficiência , Glicoproteínas de Membrana/deficiência , Regeneração Nervosa/genética , Proteínas do Tecido Nervoso/deficiência , Degeneração Retiniana/terapia , Colículos Superiores/crescimento & desenvolvimento , Vias Visuais/crescimento & desenvolvimento , Potenciais de Ação/genética , Animais , Modelos Animais de Doenças , Eletrorretinografia , Terapia Genética/tendências , Vetores Genéticos/genética , Proteínas de Filamentos Intermediários/genética , Glicoproteínas de Membrana/genética , Potenciais da Membrana/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Periferinas , Estimulação Luminosa , Células Fotorreceptoras/crescimento & desenvolvimento , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Recuperação de Função Fisiológica/genética , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Rodopsina/metabolismo , Colículos Superiores/citologia , Colículos Superiores/metabolismo , Transmissão Sináptica/genética , Regulação para Cima/genética , Visão Ocular/genética , Vias Visuais/citologia , Vias Visuais/metabolismo
17.
J Cell Biol ; 163(4): 901-10, 2003 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-14638864

RESUMO

Profilaggrin is a large epidermal polyprotein that is proteolytically processed during keratinocyte differentiation to release multiple filaggrin monomer units as well as a calcium-binding regulatory NH2-terminal filaggrin S-100 protein. We show that epidermal deficiency of the transmembrane serine protease Matriptase/MT-SP1 perturbs lipid matrix formation, cornified envelope morphogenesis, and stratum corneum desquamation. Surprisingly, proteomic analysis of Matriptase/MT-SP1-deficient epidermis revealed the selective loss of both proteolytically processed filaggrin monomer units and the NH2-terminal filaggrin S-100 regulatory protein. This was associated with a profound accumulation of profilaggrin and aberrant profilaggrin-processing products in the stratum corneum. The data identify keratinocyte Matriptase/MT-SP1 as an essential component of the profilaggrin-processing pathway and a key regulator of terminal epidermal differentiation.


Assuntos
Epiderme/enzimologia , Epiderme/crescimento & desenvolvimento , Proteínas de Filamentos Intermediários/biossíntese , Queratinócitos/enzimologia , Serina Endopeptidases/deficiência , Tripsina/deficiência , Animais , Diferenciação Celular/genética , Desidratação/enzimologia , Epiderme/patologia , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Proteínas Filagrinas , Ictiose/enzimologia , Ictiose/genética , Ictiose/patologia , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/metabolismo , Queratinócitos/patologia , Queratinócitos/ultraestrutura , Metabolismo dos Lipídeos , Proteínas de Membrana , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Peptídeo Hidrolases/deficiência , Peptídeo Hidrolases/genética , Permeabilidade , Precursores de Proteínas/metabolismo , Proteínas S100/metabolismo , Serina Endopeptidases/genética , Anormalidades da Pele/enzimologia , Anormalidades da Pele/genética , Anormalidades da Pele/patologia , Tripsina/genética
18.
J Neurochem ; 81(3): 525-32, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12065660

RESUMO

Peripherin is a type III intermediate filament (IF) abundantly expressed in developing neurons, but in the adult, it is primarily found in neurons extending to the peripheral nervous system. It has been suggested that peripherin may play a role in axonal elongation and/or cytoskeletal stabilization during development and regeneration. To further clarify the function of peripherin, we generated and characterized mice with a targeted disruption of the peripherin gene. The peripherin null mice were viable, reproduced normally and did not exhibit overt phenotypes. Microscopic analysis revealed no gross morphological defects in the ventral and dorsal roots, spinal cord, retina and gut, but protein analyses showed increased levels of the type IV IF alpha-internexin in ventral roots of peripherin null mice. Whereas the number and caliber of myelinated motor and sensory axons in the L5 roots remained unchanged in peripherin knockout mice, there was a substantial reduction ( approximately 34%) in the number of L5 unmyelinated sensory fibers that correlated with a decreased binding of the lectin IB4. These results demonstrate a requirement of peripherin for the proper development of a subset of sensory neurons.


Assuntos
Axônios/metabolismo , Proteínas de Filamentos Intermediários/deficiência , Glicoproteínas de Membrana , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/deficiência , Neurônios Aferentes/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Axônios/ultraestrutura , Contagem de Células , Imunofluorescência , Marcação de Genes , Proteínas de Filamentos Intermediários/genética , Filamentos Intermediários/ultraestrutura , Lectinas , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Neurônios Aferentes/ultraestrutura , Periferinas , RNA Mensageiro/análise
19.
J Invest Dermatol ; 115(6): 1072-81, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11121144

RESUMO

Flaky tail (gene symbol ft) is an autosomal recessive mutation in mice that results in a dry, flaky skin, and annular tail and paw constrictions in the neonatal period. Previous studies demonstrated that the ft mutation maps to the central region of mouse chromosome 3, in the vicinity of the epidermal differentiation complex, a gene locus that includes many nonkeratin genes expressed in epidermis. In this study we report a detailed characterization of the flaky tail mouse. Affected homozygous ft/ft mice exhibit large, disorganized scales on tail and paw skin, marked attenuation of the epidermal granular layer, mild acanthosis, and orthokeratotic hyperkeratosis. Biochemical analysis demonstrated that ft/ft mice lacked normal high molecular profilaggrin (approximately 500 kDa), and instead expressed a lower molecular weight form of profilaggrin (220 kDa) that is not proteolytically processed to profilaggrin intermediates or filaggrin. Mutant mice lacked the large, irregular F-type keratohyalin granules that contain profilaggrin, and filaggrin was absent from the cornified layers of ft/ft epidermis. The expression of epidermal keratins was unchanged, whereas the cornified envelope proteins involucrin and loricrin were increased in ft/ft epidermis. Cultured ft/ft keratinocytes also synthesized reduced amounts of profilaggrin mRNA and protein, demonstrating that the defect in profilaggrin expression is intrinsic to epidermal cells. These findings demonstrate that flaky tail mice express an abnormal profilaggrin polypeptide that does not form normal keratohyalin F-granules and is not proteolytically processed to filaggrin. We propose that the absence of filaggrin, and in particular the hygroscopic, filaggrin-derived amino acids that are thought to function in epidermal hydration, underlies the dry, scaly skin characteristic of ft/ft mice. This animal model provides a tool for understanding the role of filaggrin in normal epidermal function and may provide insight into the molecular basis of the filaggrin-deficient human skin disorder ichthyosis vulgaris. J Invest Dermatol 115:1072-1081 2000


Assuntos
Proteínas de Filamentos Intermediários/deficiência , Camundongos Mutantes/metabolismo , Precursores de Proteínas/deficiência , Animais , Modelos Animais de Doenças , Proteínas Filagrinas , Ictiose Vulgar/metabolismo , Proteínas de Filamentos Intermediários/biossíntese , Proteínas de Filamentos Intermediários/genética , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Peso Molecular , Mutação , Fenótipo , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , RNA Mensageiro/metabolismo , Pele/ultraestrutura
20.
J Invest Dermatol ; 114(2): 376-80, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10652001

RESUMO

We report a novel case of epidermolysis bullosa simplex with severe mucous membrane involvement and mutations in the plectin gene (PLEC1). The patient suffered from extensive blistering of the skin and oral and laryngeal mucous membranes. Electron microscopy of a lesional skin biopsy showed cleft formation within the basal cell layer of the epidermis. Antigen mapping displayed entirely negative staining for plectin, a large (>500 kDa) multifunctional adhesion protein present in hemidesmosomes of the basal keratinocytes. Mutation analysis revealed compound heterozygous, previously undisclosed nonsense mutations, Q1713X and R2351X, of paternal and maternal origin, respectively, within exon 32 of PLEC1. Based on earlier reports, plectin deficiency is associated with late onset muscular dystrophy in patients with epidermolysis bullosa. No signs of muscle weakness have been observed during the 4 y follow-up of our patient. This case illustrates the fact that molecular pathological analyses have prognostic implications in identification and evaluation of patients who appear to be at risk for development of muscular dystrophy later in life.


Assuntos
Epidermólise Bolhosa Simples/genética , Proteínas de Filamentos Intermediários/genética , Biópsia , Criança , Éxons , Humanos , Proteínas de Filamentos Intermediários/deficiência , Masculino , Mucosa , Linhagem , Plectina , Mutação Puntual , Reação em Cadeia da Polimerase , Pele/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA