Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 100(2): 364-370, 2017 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-28157540

RESUMO

SPG23 is an autosomal-recessive neurodegenerative subtype of lower limb spastic paraparesis with additional diffuse skin and hair dyspigmentation at birth followed by further patchy pigment loss during childhood. Previously, genome-wide linkage in an Arab-Israeli pedigree mapped the gene to an approximately 25 cM locus on chromosome 1q24-q32. By using whole-exome sequencing in a further Palestinian-Jordanian SPG23 pedigree, we identified a complex homozygous 4-kb deletion/20-bp insertion in DSTYK (dual serine-threonine and tyrosine protein kinase) in all four affected family members. DSTYK is located within the established linkage region and we also found the same mutation in the previously reported pedigree and another Israeli pedigree (total of ten affected individuals from three different families). The mutation removes the last two exons and part of the 3' UTR of DSTYK. Skin biopsies revealed reduced DSTYK protein levels along with focal loss of melanocytes. Ultrastructurally, swollen mitochondria and cytoplasmic vacuoles were also noted in remaining melanocytes and some keratinocytes and fibroblasts. Cultured keratinocytes and fibroblasts from an affected individual, as well as knockdown of Dstyk in mouse melanocytes, keratinocytes, and fibroblasts, were associated with increased cell death after ultraviolet irradiation. Keratinocytes from an affected individual showed loss of kinase activity upon stimulation with fibroblast growth factor. Previously, dominant mutations in DSTYK were implicated in congenital urological developmental disorders, but our study identifies different phenotypic consequences for a recurrent autosomal-recessive deletion mutation in revealing the genetic basis of SPG23.


Assuntos
Transtornos da Pigmentação/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Deleção de Sequência , Paraplegia Espástica Hereditária/genética , Vitiligo/genética , Sequência de Aminoácidos , Animais , Apoptose/genética , Povo Asiático/genética , Cromossomos Humanos Par 1/genética , Éxons , Fácies , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Ligação Genética , Loci Gênicos , Estudo de Associação Genômica Ampla , Homozigoto , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Masculino , Melanócitos/citologia , Melanócitos/metabolismo , Camundongos , Células NIH 3T3 , Linhagem , Transtornos da Pigmentação/diagnóstico , Paraplegia Espástica Hereditária/diagnóstico , Vitiligo/diagnóstico , Adulto Jovem
2.
J Am Acad Dermatol ; 83(2): 447-454, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31786163

RESUMO

BACKGROUND: Recessive dystrophic epidermolysis bullosa (RDEB) is a hereditary blistering disorder due to a lack of type VII collagen. At present, treatment is mainly supportive. OBJECTIVES: To determine whether intravenous allogeneic bone marrow-derived mesenchymal stromal/stem cells (BM-MSCs) are safe in RDEB adults and if the cells improve wound healing and quality of life. METHODS: We conducted a prospective, phase I/II, open-label study recruiting 10 RDEB adults to receive 2 intravenous infusions of BM-MSCs (on day 0 and day 14; each dose 2-4 × 106 cells/kg). RESULTS: BM-MSCs were well tolerated with no serious adverse events to 12 months. Regarding efficacy, there was a transient reduction in disease activity scores (8/10 subjects) and a significant reduction in itch. One individual showed a transient increase in type VII collagen. LIMITATIONS: Open-label trial with no placebo. CONCLUSIONS: MSC infusion is safe in RDEB adults and can have clinical benefits for at least 2 months.


Assuntos
Epidermólise Bolhosa Distrófica/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Prurido/terapia , Adolescente , Adulto , Idoso , Epidermólise Bolhosa Distrófica/complicações , Epidermólise Bolhosa Distrófica/diagnóstico , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Prurido/diagnóstico , Prurido/etiologia , Qualidade de Vida , Índice de Gravidade de Doença , Transplante Homólogo/métodos , Resultado do Tratamento , Cicatrização , Adulto Jovem
3.
Am J Hum Genet ; 95(3): 308-14, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25152456

RESUMO

Grainyhead-like 2, encoded by GRHL2, is a member of a highly conserved family of transcription factors that play essential roles during epithelial development. Haploinsufficiency for GRHL2 has been implicated in autosomal-dominant deafness, but mutations have not yet been associated with any skin pathology. We investigated two unrelated Kuwaiti families in which a total of six individuals have had lifelong ectodermal defects. The clinical features comprised nail dystrophy or nail loss, marginal palmoplantar keratoderma, hypodontia, enamel hypoplasia, oral hyperpigmentation, and dysphagia. In addition, three individuals had sensorineural deafness, and three had bronchial asthma. Taken together, the features were consistent with an unusual autosomal-recessive ectodermal dysplasia syndrome. Because of consanguinity in both families, we used whole-exome sequencing to search for novel homozygous DNA variants and found GRHL2 mutations common to both families: affected subjects in one family were homozygous for c.1192T>C (p.Tyr398His) in exon 9, and subjects in the other family were homozygous for c.1445T>A (p.Ile482Lys) in exon 11. Immortalized keratinocytes (p.Ile482Lys) showed altered cell morphology, impaired tight junctions, adhesion defects, and cytoplasmic translocation of GRHL2. Whole-skin transcriptomic analysis (p.Ile482Lys) disclosed changes in genes implicated in networks of cell-cell and cell-matrix adhesion. Our clinical findings of an autosomal-recessive ectodermal dysplasia syndrome provide insight into the role of GRHL2 in skin development, homeostasis, and human disease.


Assuntos
Fissura Palatina/genética , Proteínas de Ligação a DNA/genética , Displasia Ectodérmica/genética , Genes Recessivos/genética , Deficiência Intelectual/genética , Mutação/genética , Pele/patologia , Sindactilia/genética , Fatores de Transcrição/genética , Western Blotting , Criança , Proteínas de Ligação a DNA/metabolismo , Éxons/genética , Feminino , Humanos , Masculino , Linhagem , Fenótipo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/metabolismo , Síndrome , Fatores de Transcrição/metabolismo
4.
J Cell Sci ; 128(24): 4475-86, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26527396

RESUMO

The transmission of mechanical forces to the nucleus is important for intracellular positioning, mitosis and cell motility, yet the contribution of specific components of the cytoskeleton to nuclear mechanotransduction remains unclear. In this study, we examine how crosstalk between the cytolinker plectin and F-actin controls keratin network organisation and the 3D nuclear morphology of keratinocytes. Using micro-patterned surfaces to precisely manipulate cell shape, we find that cell adhesion and spreading regulate the size and shape of the nucleus. Disruption of the keratin cytoskeleton through loss of plectin facilitated greater nuclear deformation, which depended on acto-myosin contractility. Nuclear morphology did not depend on direct linkage of the keratin cytoskeleton with the nuclear membrane, rather loss of plectin reduced keratin filament density around the nucleus. We further demonstrate that keratinocytes have abnormal nuclear morphologies in the epidermis of plectin-deficient, epidermolysis bullosa simplex patients. Taken together, our data demonstrate that plectin is an essential regulator of nuclear morphology in vitro and in vivo and protects the nucleus from mechanical deformation.


Assuntos
Núcleo Celular/metabolismo , Mecanotransdução Celular/fisiologia , Plectina/metabolismo , Células 3T3 , Animais , Núcleo Celular/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Plectina/genética
5.
Am J Hum Genet ; 91(6): 1115-21, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23176819

RESUMO

The Rab GTPase Rab27B and one of its effector proteins, Slac2-b (also known as EXPH5, exophilin-5), have putative roles in intracellular vesicle trafficking but their relevance to human disease is not known. By using whole-exome sequencing, we identified a homozygous frameshift mutation in EXPH5 in three siblings with inherited skin fragility born to consanguineous Iraqi parents. All three individuals harbor the mutation c.5786delC (p.Pro1929Leufs(∗)8) in EXPH5, which truncates the 1,989 amino acid Slac2-b protein by 52 residues. The clinical features comprised generalized scale-crusts and occasional blisters, mostly induced by trauma, as well as mild diffuse pigmentary mottling on the trunk and proximal limbs. There was no increased bleeding tendency, no neurologic abnormalities, and no increased incidence of infection. Analysis of an affected person's skin showed loss of Slac2-b immunostaining (C-terminal antibody), disruption of keratinocyte adhesion within the lower epidermis, and an increased number of perinuclear vesicles. A role for Slac2-b in keratinocyte biology was supported by findings of cytoskeletal disruption (mainly keratin intermediate filaments) and decreased keratinocyte adhesion in both keratinocytes from an affected subject and after shRNA knockdown of Slac2-b in normal keratinocytes. Slac2-b was also shown to colocalize with Rab27B and ß4 integrin to early adhesion initiation sites in spreading normal keratinocytes. Collectively, our findings identify an unexpected role for Slac2-b in inherited skin fragility and expand the clinical spectrum of human disorders of GTPase effector proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Mutação em Linhagem Germinativa , Doenças do Cabelo/congênito , Doenças do Cabelo/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Bases , Feminino , Doenças do Cabelo/diagnóstico , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Masculino , Linhagem , Pele/patologia , Pele/ultraestrutura , Proteínas rab de Ligação ao GTP/metabolismo
6.
Nat Med ; 13(3): 378-83, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17322897

RESUMO

Transmissibility of characteristic lesions to experimental animals may help us understand the pathomechanism of human autoimmune disease. Here we show that human autoimmune disease can be reproduced using genetically engineered model mice. Bullous pemphigoid (BP) is the most common serious autoimmune blistering skin disease, with a considerable body of indirect evidence indicating that the underlying autoantigen is collagen XVII (COL17). Passive transfer of human BP autoantibodies into mice does not induce skin lesions, probably because of differences between humans and mice in the amino acid sequence of the COL17 pathogenic epitope. We injected human BP autoantibody into Col17-knockout mice rescued by the human ortholog. This resulted in BP-like skin lesions and a human disease phenotype. Humanization of autoantigens is a new approach to the study of human autoimmune diseases.


Assuntos
Autoantígenos/química , Colágenos não Fibrilares/genética , Penfigoide Bolhoso/imunologia , Animais , Autoanticorpos/fisiologia , Autoantígenos/genética , Autoantígenos/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Colágenos não Fibrilares/deficiência , Colágeno Tipo XVII
7.
J Am Acad Dermatol ; 68(3): 482-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23219556

RESUMO

BACKGROUND: Diffuse melanosis cutis (DMC) is a rare presentation of metastatic melanoma characterized by a progressive blue-gray discoloration of the skin and mucous membranes. OBJECTIVE: To foster a better understanding of the clinical presentation, histological findings, and pathophysiology underlying DMC. METHODS: A systematic review of the literature was completed utilizing MEDLINE, CINAHL, Embase, and Google. Data were extracted using a protocol-driven spread sheet with all statistical analyses completed using SPSS. RESULTS: The review identified 68 original cases of DMC. The mean time from diagnosis of melanoma until development of DMC was 11.48 months (95% confidence interval [CI]: 0-48.16). The mean time to death following the onset of DMC was 4.43 months (95% CI: 0.00-11.11). Histological findings were relatively consistent demonstrating intracellular and extracellular melanin deposition in the dermis, with a pronounced perivascular distribution. The pathophysiological mechanisms underlying DMC could not be definitively elucidated; however, it is hypothesized that the melanin precursors, melanin, and melanosomes liberated by cytolytic metastatic melanoma deposits are phagocytosed by dermal histiocytes, manifesting clinically as diffuse melanosis. LIMITATIONS: The cross-sectional nature of case reports, paucity of cases of DMC, and heterogeneity in reporting limit any conclusions being drawn regarding the pathophysiology of DMC definitively. CONCLUSION: DMC heralds a poor prognosis for patients with metastatic melanoma and affected patients should be made aware of the implications of this condition on survival.


Assuntos
Melanoma/secundário , Melanose/patologia , Neoplasias Cutâneas/patologia , Pigmentação da Pele , Pele/patologia , Derme/patologia , Humanos , Melaninas/metabolismo , Melanoma/patologia , Melanossomas/patologia , Prognóstico
8.
Proc Natl Acad Sci U S A ; 107(32): 14345-50, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20660747

RESUMO

Attempts to treat congenital protein deficiencies using bone marrow-derived cells have been reported. These efforts have been based on the concepts of stem cell plasticity. However, it is considered more difficult to restore structural proteins than to restore secretory enzymes. This study aims to clarify whether bone marrow transplantation (BMT) treatment can rescue epidermolysis bullosa (EB) caused by defects in keratinocyte structural proteins. BMT treatment of adult collagen XVII (Col17) knockout mice induced donor-derived keratinocytes and Col17 expression associated with the recovery of hemidesmosomal structure and better skin manifestations, as well improving the survival rate. Both hematopoietic and mesenchymal stem cells have the potential to produce Col17 in the BMT treatment model. Furthermore, human cord blood CD34(+) cells also differentiated into keratinocytes and expressed human skin component proteins in transplanted immunocompromised (NOD/SCID/gamma(c)(null)) mice. The current conventional BMT techniques have significant potential as a systemic therapeutic approach for the treatment of human EB.


Assuntos
Membrana Basal/metabolismo , Transplante de Medula Óssea/fisiologia , Epidermólise Bolhosa/terapia , Proteínas de Membrana/biossíntese , Animais , Autoantígenos/biossíntese , Membrana Basal/química , Epiderme , Humanos , Queratinócitos/citologia , Camundongos , Camundongos SCID , Colágenos não Fibrilares/biossíntese , Colágenos não Fibrilares/deficiência , Transplante de Células-Tronco , Taxa de Sobrevida , Resultado do Tratamento , Colágeno Tipo XVII
9.
Am J Pathol ; 176(2): 914-25, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20042683

RESUMO

Activation of the complement cascade via the classical pathway is required for the development of tissue injury in many autoantibody-mediated diseases. It therefore makes sense to block the pathological action of autoantibodies by preventing complement activation through inhibition of autoantibody binding to the corresponding pathogenic autoantigen using targeted Fab antibody fragments. To achieve this, we use bullous pemphigoid (BP) as an example of a typical autoimmune disease. Recombinant Fabs against the non-collagenous 16th-A domain of type XVII collagen, the main pathogenic epitope for autoantibodies in BP, were generated from antibody repertoires of BP patients by phage display. Two Fabs, Fab-B4 and Fab-19, showed marked ability to inhibit the binding of BP autoantibodies and subsequent complement activation in vitro. In the in vivo experiments using type XVII collagen humanized BP model mice, these Fabs protected mice against BP autoantibody-induced blistering disease. Thus, the blocking of pathogenic epitopes using engineered Fabs appears to demonstrate efficacy and may lead to disease-specific treatments for antibody-mediated autoimmune diseases.


Assuntos
Autoanticorpos/metabolismo , Autoantígenos/imunologia , Doenças Autoimunes/terapia , Fragmentos Fab das Imunoglobulinas/farmacologia , Animais , Animais Recém-Nascidos , Autoanticorpos/efeitos adversos , Autoanticorpos/imunologia , Autoanticorpos/isolamento & purificação , Autoantígenos/efeitos dos fármacos , Doenças Autoimunes/etiologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Imunoterapia/métodos , Camundongos , Camundongos Nus , Penfigoide Bolhoso/sangue , Penfigoide Bolhoso/imunologia , Penfigoide Bolhoso/patologia , Penfigoide Bolhoso/terapia , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico
10.
Hum Mutat ; 31(3): 308-16, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20052759

RESUMO

Plectin is a cytoskeletal linker protein that has a dumbbell-like structure with a long central rod and N- and C-terminal globular domains. Mutations in the gene encoding plectin (PLEC1) cause two distinct autosomal recessive subtypes of epidermolysis bullosa (EB): EB simplex with muscular dystrophy (EBS-MD), and EB simplex with pyloric atresia (EBS-PA). Here, we demonstrate that normal human fibroblasts express two different plectin isoforms including full-length and rodless forms of plectin. We performed detailed analysis of plectin expression patterns in six EBS-MD and three EBS-PA patients. In EBS-PA, expression of all plectin domains was found to be markedly attenuated or completely lost; in EBS-MD, the expression of the N- and C-terminal domains of plectin remained detectable, although the expression of rod domains was absent or markedly reduced. Our data suggest that loss of the full-length plectin isoform with residual expression of the rodless plectin isoform leads to EBS-MD, and that complete loss or marked attenuation of full-length and rodless plectin expression underlies the more severe EBS-PA phenotype. These results also clearly account for the majority of EBS-MD PLEC1 mutation restriction within the large exon 31 that encodes the plectin rod domain, whereas EBS-PA PLEC1 mutations are generally outside exon 31.


Assuntos
Epidermólise Bolhosa Simples/metabolismo , Plectina/metabolismo , Algoritmos , Análise Mutacional de DNA , Epidermólise Bolhosa Simples/diagnóstico , Éxons , Fibroblastos/metabolismo , Humanos , Queratinócitos/citologia , Microscopia de Fluorescência/métodos , Modelos Genéticos , Músculo Esquelético/metabolismo , Mutação , Plectina/química , Isoformas de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA