Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
2.
Nature ; 491(7425): 603-7, 2012 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-23075850

RESUMO

Nuclear-architecture defects have been shown to correlate with the manifestation of a number of human diseases as well as ageing. It is therefore plausible that diseases whose manifestations correlate with ageing might be connected to the appearance of nuclear aberrations over time. We decided to evaluate nuclear organization in the context of ageing-associated disorders by focusing on a leucine-rich repeat kinase 2 (LRRK2) dominant mutation (G2019S; glycine-to-serine substitution at amino acid 2019), which is associated with familial and sporadic Parkinson's disease as well as impairment of adult neurogenesis in mice. Here we report on the generation of induced pluripotent stem cells (iPSCs) derived from Parkinson's disease patients and the implications of LRRK2(G2019S) mutation in human neural-stem-cell (NSC) populations. Mutant NSCs showed increased susceptibility to proteasomal stress as well as passage-dependent deficiencies in nuclear-envelope organization, clonal expansion and neuronal differentiation. Disease phenotypes were rescued by targeted correction of the LRRK2(G2019S) mutation with its wild-type counterpart in Parkinson's disease iPSCs and were recapitulated after targeted knock-in of the LRRK2(G2019S) mutation in human embryonic stem cells. Analysis of human brain tissue showed nuclear-envelope impairment in clinically diagnosed Parkinson's disease patients. Together, our results identify the nucleus as a previously unknown cellular organelle in Parkinson's disease pathology and may help to open new avenues for Parkinson's disease diagnoses as well as for the potential development of therapeutics targeting this fundamental cell structure.


Assuntos
Proteínas Mutantes/metabolismo , Células-Tronco Neurais/patologia , Doença de Parkinson/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Apoptose , Diferenciação Celular , Divisão Celular , Linhagem Celular , Células Clonais/metabolismo , Células Clonais/patologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Técnicas de Introdução de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Proteínas Mutantes/genética , Mutação , Células-Tronco Neurais/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Estresse Fisiológico
3.
Nat Methods ; 10(1): 77-83, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23202434

RESUMO

Lineage conversion of one somatic cell type to another is an attractive approach for generating specific human cell types. Lineage conversion can be direct, in the absence of proliferation and multipotent progenitor generation, or indirect, by the generation of expandable multipotent progenitor states. We report the development of a reprogramming methodology in which cells transition through a plastic intermediate state, induced by brief exposure to reprogramming factors, followed by differentiation. We use this approach to convert human fibroblasts to mesodermal progenitor cells, including by non-integrative approaches. These progenitor cells demonstrated bipotent differentiation potential and could generate endothelial and smooth muscle lineages. Differentiated endothelial cells exhibited neo-angiogenesis and anastomosis in vivo. This methodology for indirect lineage conversion to angioblast-like cells adds to the armamentarium of reprogramming approaches aimed at the study and treatment of ischemic pathologies.


Assuntos
Diferenciação Celular , Linhagem da Célula , Reprogramação Celular , Endotélio Vascular/citologia , Fibroblastos/citologia , Miócitos de Músculo Liso/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Western Blotting , Movimento Celular , Proliferação de Células , Células Cultivadas , Endotélio Vascular/metabolismo , Fibroblastos/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Camundongos , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo
4.
Stem Cells ; 32(11): 2923-2938, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25175072

RESUMO

Reprogramming technologies have emerged as a promising approach for future regenerative medicine. Here, we report on the establishment of a novel methodology allowing for the conversion of human fibroblasts into hematopoietic progenitor-like cells with macrophage differentiation potential. SOX2 overexpression in human fibroblasts, a gene found to be upregulated during hematopoietic reconstitution in mice, induced the rapid appearance of CD34+ cells with a concomitant upregulation of mesoderm-related markers. Profiling of cord blood hematopoietic progenitor cell populations identified miR-125b as a factor facilitating commitment of SOX2-generated CD34+ cells to immature hematopoietic-like progenitor cells with grafting potential. Further differentiation toward the monocytic lineage resulted in the appearance of CD14+ cells with functional phagocytic capacity. In vivo transplantation of SOX2/miR-125b-generated CD34+ cells facilitated the maturation of the engrafted cells toward CD45+ cells and ultimately the monocytic/macrophage lineage. Altogether, our results indicate that strategies combining lineage conversion and further lineage specification by in vivo or in vitro approaches could help to circumvent long-standing obstacles for the reprogramming of human cells into hematopoietic cells with clinical potential.


Assuntos
Diferenciação Celular/fisiologia , Fibroblastos/citologia , Monócitos/citologia , Células-Tronco/citologia , Animais , Antígenos CD34/metabolismo , Linhagem da Célula/fisiologia , Células Cultivadas , Humanos , Antígenos Comuns de Leucócito/metabolismo , Camundongos
5.
Proc Natl Acad Sci U S A ; 105(32): 11242-7, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18678914

RESUMO

Organ shape and size, and, ultimately, organ function, relate in part to the cell and tissue spatial arrangement that takes place during embryonic development. Despite great advances in the genetic regulatory networks responsible for tissue and organ development, it is not yet clearly understood how specific gene functions are linked to the specific morphogenetic processes underlying the internal organ asymmetries found in vertebrate animals. During female chick embryogenesis, and in contrast to males where both testes develop symmetrically, asymmetrical gonad morphogenesis results in only one functional ovary. The disposition of paired organs along the left-right body axis has been shown to be regulated by the activity of the homeobox containing gene pitx2. We have found that pitx2 regulates cell adhesion, affinity, and cell recognition events in the developing gonad primordium epithelia. This in turn not only allows for proper somatic development of the gonad cortex but also permits the proliferation and differentiation of primordial germ cells. We illustrate how Pitx2 activity directs asymmetrical gonad morphogenesis by controlling mitotic spindle orientation of the developing gonad cortex and how, by modulating cyclinD1 expression during asymmetric ovarian development, Pitx2 appears to control gonad organ size. All together our observations indicate that the effects elicited by Pitx2 during the development of the female chick ovary are critical for cell topology, growth, fate, and ultimately organ morphogenesis and function.


Assuntos
Diferenciação Celular/fisiologia , Galinhas/fisiologia , Células Germinativas/fisiologia , Ovário/embriologia , Animais , Adesão Celular/fisiologia , Proliferação de Células , Embrião de Galinha , Ciclina D1/metabolismo , Epitélio/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Germinativas/citologia , Proteínas de Homeodomínio , Masculino , Tamanho do Órgão , Ovário/citologia , Fuso Acromático/metabolismo , Testículo/citologia , Testículo/embriologia , Fatores de Transcrição , Proteína Homeobox PITX2
6.
Mol Cell Biol ; 24(12): 5257-68, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15169890

RESUMO

Although largely involved in innate and adaptive immunity, NF-kappa B plays an important role in vertebrate development. In chicks, the inactivation of the NF-kappa B pathway induces functional alterations of the apical ectodermal ridge, which mediates limb outgrowth. In mice, the complete absence of NF-kappa B activity leads to prenatal death and neural tube defects. Here, we report the cloning and characterization of NF-kappa B/I kappa B proteins in zebra fish. Despite being ubiquitously expressed among the embryonic tissues, NF-kappa B/I kappa B members present distinct patterns of gene expression during the early zebra fish development. Biochemical assays indicate that zebra fish NF-kappa B proteins are able to bind consensus DNA-binding (kappa B) sites and inhibitory I kappa B alpha proteins from mammals. We show that zebra fish I kappa B alphas are degraded in a time-dependent manner after induction of transduced murine embryo fibroblasts (MEFs) and that these proteins are able to rescue NF-kappa B activity in I kappa B alpha(-/-) MEFs. Expression of a dominant-negative form of the murine I kappa B alpha (mI kappa B alpha M), which is able to block NF-kappa B in zebra fish cells, interferes with the notochord differentiation, generating no tail (ntl)-like embryos. This phenotype can be rescued by coinjection of the T-box gene ntl (Brachyury homologue), which is typically required for the formation of posterior mesoderm and axial development, suggesting that ntl lies downstream of NF-kappa B . We further show that ntl and Brachyury promoter regions contain functional kappa B sites and NF-kappa B can directly modulate ntl expression. Our study illustrates the conservation and compatibility of NF-kappa B/I kappa B proteins among vertebrates and the importance of NF-kappa B pathway in mesoderm formation during early embryogenesis.


Assuntos
Proteínas I-kappa B/metabolismo , NF-kappa B/metabolismo , Notocorda/embriologia , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Células Cultivadas , DNA Complementar/genética , Proteínas Fetais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Proteínas I-kappa B/genética , Camundongos , Camundongos Knockout , Inibidor de NF-kappaB alfa , NF-kappa B/genética , Notocorda/metabolismo , Fenótipo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Nat Commun ; 7: 10743, 2016 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-26899176

RESUMO

Glioma tumour-initiating cells (GTICs) can originate upon the transformation of neural progenitor cells (NPCs). Studies on GTICs have focused on primary tumours from which GTICs could be isolated and the use of human embryonic material. Recently, the somatic genomic landscape of human gliomas has been reported. RTK (receptor tyrosine kinase) and p53 signalling were found dysregulated in ∼90% and 86% of all primary tumours analysed, respectively. Here we report on the use of human-induced pluripotent stem cells (hiPSCs) for modelling gliomagenesis. Dysregulation of RTK and p53 signalling in hiPSC-derived NPCs (iNPCs) recapitulates GTIC properties in vitro. In vivo transplantation of transformed iNPCs leads to highly aggressive tumours containing undifferentiated stem cells and their differentiated derivatives. Metabolic modulation compromises GTIC viability. Last, screening of 101 anti-cancer compounds identifies three molecules specifically targeting transformed iNPCs and primary GTICs. Together, our results highlight the potential of hiPSCs for studying human tumourigenesis.


Assuntos
Transformação Celular Neoplásica , Glioma/etiologia , Células-Tronco Pluripotentes Induzidas , Células-Tronco Neoplásicas/fisiologia , Células-Tronco Neurais/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Fatores de Transcrição SOXB1/metabolismo , Ensaio Tumoral de Célula-Tronco
8.
Nat Commun ; 5: 4330, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24999918

RESUMO

Fanconi anaemia (FA) is a recessive disorder characterized by genomic instability, congenital abnormalities, cancer predisposition and bone marrow (BM) failure. However, the pathogenesis of FA is not fully understood partly due to the limitations of current disease models. Here, we derive integration free-induced pluripotent stem cells (iPSCs) from an FA patient without genetic complementation and report in situ gene correction in FA-iPSCs as well as the generation of isogenic FANCA-deficient human embryonic stem cell (ESC) lines. FA cellular phenotypes are recapitulated in iPSCs/ESCs and their adult stem/progenitor cell derivatives. By using isogenic pathogenic mutation-free controls as well as cellular and genomic tools, our model serves to facilitate the discovery of novel disease features. We validate our model as a drug-screening platform by identifying several compounds that improve hematopoietic differentiation of FA-iPSCs. These compounds are also able to rescue the hematopoietic phenotype of FA patient BM cells.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Anemia de Fanconi/etiologia , Anemia de Fanconi/patologia , Modelos Biológicos , Células-Tronco/patologia , Diferenciação Celular , Epigênese Genética , Anemia de Fanconi/tratamento farmacológico , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Adulto Jovem
9.
Nat Cell Biol ; 15(12): 1507-15, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24240476

RESUMO

Diseases affecting the kidney constitute a major health issue worldwide. Their incidence and poor prognosis affirm the urgent need for the development of new therapeutic strategies. Recently, differentiation of pluripotent cells to somatic lineages has emerged as a promising approach for disease modelling and cell transplantation. Unfortunately, differentiation of pluripotent cells into renal lineages has demonstrated limited success. Here we report on the differentiation of human pluripotent cells into ureteric-bud-committed renal progenitor-like cells. The generated cells demonstrated rapid and specific expression of renal progenitor markers on 4-day exposure to defined media conditions. Further maturation into ureteric bud structures was accomplished on establishment of a three-dimensional culture system in which differentiated human cells assembled and integrated alongside murine cells for the formation of chimeric ureteric buds. Altogether, our results provide a new platform for the study of kidney diseases and lineage commitment, and open new avenues for the future application of regenerative strategies in the clinic.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/fisiologia , Rim/patologia , Animais , Técnicas de Cultura de Células , Células-Tronco Embrionárias/fisiologia , Humanos , Células MCF-7 , Mesoderma/patologia , Camundongos , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/terapia , Medicina Regenerativa , Transplante de Células-Tronco , Técnicas de Cultura de Tecidos , Tretinoína/fisiologia
10.
Cell Stem Cell ; 8(6): 688-94, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21596650

RESUMO

Combination of stem cell-based approaches with gene-editing technologies represents an attractive strategy for studying human disease and developing therapies. However, gene-editing methodologies described to date for human cells suffer from technical limitations including limited target gene size, low targeting efficiency at transcriptionally inactive loci, and off-target genetic effects that could hamper broad clinical application. To address these limitations, and as a proof of principle, we focused on homologous recombination-based gene correction of multiple mutations on lamin A (LMNA), which are associated with various degenerative diseases. We show that helper-dependent adenoviral vectors (HDAdVs) provide a highly efficient and safe method for correcting mutations in large genomic regions in human induced pluripotent stem cells and can also be effective in adult human mesenchymal stem cells. This type of approach could be used to generate genotype-matched cell lines for disease modeling and drug discovery and potentially also in therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Lamina Tipo A/genética , Mutação , Linhagem Celular , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia
11.
Genes Dev ; 20(23): 3232-7, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17114576

RESUMO

The cellular and molecular bases allowing tissue regeneration are not well understood. By performing gain- and loss-of-function experiments of specific members of the Wnt pathway during appendage regeneration, we demonstrate that this pathway is not only necessary for regeneration to occur, but it is also able to promote regeneration in axolotl, Xenopus, and zebrafish. Furthermore, we show that changes in the spatiotemporal distribution of beta-catenin in the developing chick embryo elicit apical ectodermal ridge and limb regeneration in an organism previously thought not to regenerate. Our studies may provide valuable insights toward a better understanding of adult tissue regeneration.


Assuntos
Extremidades/fisiologia , Regeneração/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Ambystoma/crescimento & desenvolvimento , Animais , Larva , Xenopus , Proteínas de Xenopus/fisiologia , Peixe-Zebra
13.
Genes Dev ; 17(10): 1213-8, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12730123

RESUMO

Left-sided expression of Nodal in the lateral plate mesoderm is a conserved feature necessary for the establishment of normal left-right asymmetry during vertebrate embryogenesis. By using gain- and loss-of-function experiments in zebrafish and mouse, we show that the activity of the Notch pathway is necessary and sufficient for Nodal expression around the node, and for proper left-right determination. We identify Notch-responsive elements in the Nodal promoter, and unveil a direct relationship between Notch activity and Nodal expression around the node. Our findings provide evidence for a mechanism involving Notch activity that translates an initial symmetry-breaking event into asymmetric gene expression.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/fisiologia , Fator de Crescimento Transformador beta/genética , Animais , Elementos Facilitadores Genéticos , Proteínas Hedgehog , Camundongos , Proteína Nodal , Organizadores Embrionários/fisiologia , Regiões Promotoras Genéticas , Receptores Notch , Transdução de Sinais/fisiologia , Situs Inversus/embriologia , Transativadores/genética , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA