Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Lymphology ; 55(3): 129-134, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36446400

RESUMO

We have studied the lymphatic phenotypes of 2 mutations, known to cause abnormalities of lymphatics in humans, in mice. The Cx47 R260C mutation (variably penetrant in humans heterozygous for it and causing limb lymphedema) had an adult mouse phenotype of hyperplasia and increased lymph nodes only in homozygous condition but we did not find any anatomical phenotype in day 16.5 homozygous embryos. Mice harboring the Sos1 mutation E846K (causing Noonan's in man which occasionally shows lymphatic dysplasia) had no adult heterozygous phenotype in lymphatic vessel appearance and drainage (homozygotes are early embryonic lethals) while day 16.5 heterozygous embryos also had no detectable anatomical phenotype.


Assuntos
Doenças Linfáticas , Vasos Linfáticos , Proteína SOS1 , Animais , Humanos , Camundongos , Heterozigoto , Homozigoto , Vasos Linfáticos/anormalidades , Vasos Linfáticos/embriologia , Mutação , Fenótipo , Linfedema/embriologia , Linfedema/genética , Doenças Linfáticas/embriologia , Doenças Linfáticas/genética , Proteína SOS1/genética , Conexinas/genética
2.
Dev Biol ; 409(1): 218-233, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26542011

RESUMO

Lymph is returned to the blood circulation exclusively via four lymphovenous valves (LVVs). Despite their vital importance, the architecture and development of LVVs is poorly understood. We analyzed the formation of LVVs at the molecular and ultrastructural levels during mouse embryogenesis and identified three critical steps. First, LVV-forming endothelial cells (LVV-ECs) differentiate from PROX1(+) progenitors and delaminate from the luminal side of the veins. Second, LVV-ECs aggregate, align perpendicular to the direction of lymph flow and establish lympho-venous connections. Finally, LVVs mature with the recruitment of mural cells. LVV morphogenesis is disrupted in four different mouse models of primary lymphedema and the severity of LVV defects correlate with that of lymphedema. In summary, we have provided the first and the most comprehensive analysis of LVV development. Furthermore, our work suggests that aberrant LVVs contribute to lymphedema.


Assuntos
Vasos Linfáticos/embriologia , Linfedema/embriologia , Linfedema/patologia , Válvulas Venosas/embriologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Células Endoteliais/ultraestrutura , Vasos Linfáticos/ultraestrutura , Camundongos Endogâmicos C57BL , Morfogênese , Penetrância , Fenótipo , Válvulas Venosas/ultraestrutura
3.
Cell Reprogram ; 17(4): 275-87, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26474030

RESUMO

The generation of human induced pluripotent stem cells (hiPSCs) derived from an autologous extraembryonic fetal source is an innovative personalized regenerative technology that can transform own-self cells into embryonic stem-like ones. These cells are regarded as a promising candidate for cell-based therapy, as well as an ideal target for disease modeling and drug discovery. Thus, hiPSCs enable researchers to undertake studies for treating diseases or for future applications of in utero therapy. We used a polycistronic lentiviral vector (hSTEMCCA-loxP) encoding OCT4, SOX2, KLF4, and cMYC genes and containing loxP sites, excisible by Cre recombinase, to reprogram patient-specific fetal cells derived from prenatal diagnosis for several genetic disorders, such as myotonic dystrophy type 1 (DM1), ß-thalassemia (ß-Thal), lymphedema-distichiasis syndrome (LDS), spinal muscular atrophy (SMA), cystic fibrosis (CF), as well as from wild-type (WT) fetal cells. Because cell types tested to create hiPSCs influence both the reprogramming process efficiency and the kinetics, we used chorionic villus (CV) and amniotic fluid (AF) cells, demonstrating how they represent an ideal cell resource for a more efficient generation of hiPSCs. The successful reprogramming of both CV and AF cells into hiPSCs was confirmed by specific morphological, molecular, and immunocytochemical markers and also by their teratogenic potential when inoculated in vivo. We further demonstrated the stability of reprogrammed cells over 10 and more passages and their capability to differentiate into the three embryonic germ layers, as well as into neural cells. These data suggest that hiPSCs-CV/AF can be considered a valid cellular model to accomplish pathogenesis studies and therapeutic applications.


Assuntos
Feto/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Células Cultivadas , Reprogramação Celular , Amostra da Vilosidade Coriônica , Fibrose Cística/embriologia , Pestanas/anormalidades , Pestanas/embriologia , Feminino , Feto/fisiologia , Vetores Genéticos , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Lentivirus/genética , Linfedema/embriologia , Camundongos , Atrofia Muscular Espinal/embriologia , Distrofia Miotônica/embriologia , Fator 3 de Transcrição de Octâmero/genética , Gravidez , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição SOXB1/genética , Transgenes , Talassemia beta/embriologia
4.
J Clin Invest ; 125(8): 2924-6, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26214518

RESUMO

Hemodynamic forces regulate many aspects of blood vessel disease and development, including susceptibility to atherosclerosis and remodeling of primary blood vessels into a mature vascular network. Vessels of the lymphatic circulatory system are also subjected to fluid flow-associated forces, but the molecular and cellular mechanisms by which these forces regulate the formation and maintenance of lymphatic vessels remain largely uncharacterized. This issue of the JCI includes two articles that begin to address how fluid flow influences lymphatic vessel development and function. Sweet et al. demonstrate that lymph flow is essential for the remodeling of primary lymphatic vessels, for ensuring the proper distribution of smooth muscle cells (SMCs), and for the development and maturation of lymphatic valves. Kazenwadel et al. show that flow-induced lymphatic valve development is initiated by the upregulation of GATA2, which has been linked to lymphedema in patients with Emberger syndrome. Together, these observations and future studies inspired by these results have potential to lead to the development of strategies for the treatment of lymphatic disorders.


Assuntos
Fator de Transcrição GATA2/metabolismo , Linfa/fisiologia , Vasos Linfáticos/embriologia , Linfedema/embriologia , Músculo Liso Vascular/embriologia , Mutação , Miócitos de Músculo Liso/metabolismo , Animais , Humanos
5.
J Clin Invest ; 125(8): 2979-94, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26214525

RESUMO

Heterozygous germline mutations in the zinc finger transcription factor GATA2 have recently been shown to underlie a range of clinical phenotypes, including Emberger syndrome, a disorder characterized by lymphedema and predisposition to myelodysplastic syndrome/acute myeloid leukemia (MDS/AML). Despite well-defined roles in hematopoiesis, the functions of GATA2 in the lymphatic vasculature and the mechanisms by which GATA2 mutations result in lymphedema have not been characterized. Here, we have provided a molecular explanation for lymphedema predisposition in a subset of patients with germline GATA2 mutations. Specifically, we demonstrated that Emberger-associated GATA2 missense mutations result in complete loss of GATA2 function, with respect to the capacity to regulate the transcription of genes that are important for lymphatic vessel valve development. We identified a putative enhancer element upstream of the key lymphatic transcriptional regulator PROX1 that is bound by GATA2, and the transcription factors FOXC2 and NFATC1. Emberger GATA2 missense mutants had a profoundly reduced capacity to bind this element. Conditional Gata2 deletion in mice revealed that GATA2 is required for both development and maintenance of lymphovenous and lymphatic vessel valves. Together, our data unveil essential roles for GATA2 in the lymphatic vasculature and explain why a select catalogue of human GATA2 mutations results in lymphedema.


Assuntos
Fator de Transcrição GATA2/metabolismo , Vasos Linfáticos/embriologia , Linfedema/embriologia , Mutação , Animais , Elementos Facilitadores Genéticos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição GATA2/genética , Deleção de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células K562 , Vasos Linfáticos/patologia , Linfedema/genética , Linfedema/patologia , Camundongos , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
6.
Hum Mol Genet ; 18(15): 2839-50, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19429912

RESUMO

Mutations in the transcription factor gene SOX18 cause vascular, lymphatic and hair follicle defects in humans with dominant and recessive forms of hypotrichosis-lymphedema-telangiectasia (HLT) syndrome. Here, we clarify the role of SOX18 in the vascular dysfunction in HLT by ultrastructural, immunofluorescence, molecular and functional analysis of vascular anomalies in embryos of the naturally occurring Sox18-mutant mouse strain ragged-opossum (Ra(Op)). Early genesis and patterning of vasculature was unimpaired in Ra(Op) embryos, but surface capillaries became enlarged from 12.5 dpc and embryos developed massive surface hemorrhage by 14.5 dpc. Large focal breaches in the endothelial barrier were observed, in addition to endothelial hyperplasia associated with impaired pericyte recruitment to the microvasculature. Expression of the genes encoding the endothelial factors MMP7, IL7R and N-cadherin was reduced in Ra(Op) embryos, suggesting that these are downstream targets of SOX18. Together, our results indicate that vascular anomalies in HLT arise from defects in regulation of genes required for the acquisition of structural integrity during microvascular maturation.


Assuntos
Vasos Sanguíneos/fisiopatologia , Hipotricose/metabolismo , Linfedema/metabolismo , Fatores de Transcrição SOXF/metabolismo , Telangiectasia/metabolismo , Animais , Vasos Sanguíneos/anormalidades , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Modelos Animais de Doenças , Humanos , Hipotricose/embriologia , Hipotricose/genética , Hipotricose/fisiopatologia , Linfedema/embriologia , Linfedema/genética , Linfedema/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Fatores de Transcrição SOXF/genética , Telangiectasia/embriologia , Telangiectasia/genética , Telangiectasia/fisiopatologia
7.
Lymphology ; 11(1): 10-21, 1978 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-642583

RESUMO

The morphogenesis of congenital hereditary lymphedema was studied in pigs. The disorder, which is essentially a general underdevelopment or even total non-development of the lymphatic system proved to be present during the whole period of lymphatic development. It is suggested that a retardation in the differentiation of the lymphatic primordia from the primitive veins is the early event regulated by a chromosomal aberration. The longer this delay the more serious the lymphatic malformations.


Assuntos
Linfedema/veterinária , Doenças dos Suínos/embriologia , Animais , Sistema Linfático/embriologia , Linfedema/congênito , Linfedema/embriologia , Linfedema/patologia , Suínos , Doenças dos Suínos/congênito , Doenças dos Suínos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA