Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep Med ; 5(3): 101449, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38508141

RESUMO

Tissue regeneration following an injury requires dynamic cell-state transitions that allow for establishing the cell identities required for the restoration of tissue homeostasis and function. Here, we present a biochemical intervention that induces an intermediate cell state mirroring a transition identified during normal differentiation of myoblasts and other multipotent and pluripotent cells to mature cells. When applied in somatic differentiated cells, the intervention, composed of one-carbon metabolites, reduces some dedifferentiation markers without losing the lineage identity, thus inducing limited reprogramming into a more flexible cell state. Moreover, the intervention enabled accelerated repair after muscle injury in young and aged mice. Overall, our study uncovers a conserved biochemical transitional phase that enhances cellular plasticity in vivo and hints at potential and scalable biochemical interventions of use in regenerative medicine and rejuvenation interventions that may be more tractable than genetic ones.


Assuntos
Músculos , Mioblastos , Camundongos , Animais , Diferenciação Celular , Mioblastos/metabolismo
2.
Cell Stem Cell ; 31(1): 52-70.e8, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-38181751

RESUMO

Human pluripotent stem cell-derived kidney organoids offer unprecedented opportunities for studying polycystic kidney disease (PKD), which still has no effective cure. Here, we developed both in vitro and in vivo organoid models of PKD that manifested tubular injury and aberrant upregulation of renin-angiotensin aldosterone system. Single-cell analysis revealed that a myriad of metabolic changes occurred during cystogenesis, including defective autophagy. Experimental activation of autophagy via ATG5 overexpression or primary cilia ablation significantly inhibited cystogenesis in PKD kidney organoids. Employing the organoid xenograft model of PKD, which spontaneously developed tubular cysts, we demonstrate that minoxidil, a potent autophagy activator and an FDA-approved drug, effectively attenuated cyst formation in vivo. This in vivo organoid model of PKD will enhance our capability to discover novel disease mechanisms and validate candidate drugs for clinical translation.


Assuntos
Cílios , Doenças Renais Policísticas , Humanos , Rim , Doenças Renais Policísticas/tratamento farmacológico , Autofagia , Organoides
3.
Sci Transl Med ; 14(657): eabl6057, 2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-35947677

RESUMO

Constitutive heterochromatin is responsible for genome repression of DNA enriched in repetitive sequences, telomeres, and centromeres. During physiological and pathological premature aging, heterochromatin homeostasis is profoundly compromised. Here, we showed that LINE-1 (Long Interspersed Nuclear Element-1; L1) RNA accumulation was an early event in both typical and atypical human progeroid syndromes. L1 RNA negatively regulated the enzymatic activity of the histone-lysine N-methyltransferase SUV39H1 (suppression of variegation 3-9 homolog 1), resulting in heterochromatin loss and onset of senescent phenotypes in vitro. Depletion of L1 RNA in dermal fibroblast cells from patients with different progeroid syndromes using specific antisense oligonucleotides (ASOs) restored heterochromatin histone 3 lysine 9 and histone 3 lysine 27 trimethylation marks, reversed DNA methylation age, and counteracted the expression of senescence-associated secretory phenotype genes such as p16, p21, activating transcription factor 3 (ATF3), matrix metallopeptidase 13 (MMP13), interleukin 1a (IL1a), BTG anti-proliferation factor 2 (BTG2), and growth arrest and DNA damage inducible beta (GADD45b). Moreover, systemic delivery of ASOs rescued the histophysiology of tissues and increased the life span of a Hutchinson-Gilford progeria syndrome mouse model. Transcriptional profiling of human and mouse samples after L1 RNA depletion demonstrated that pathways associated with nuclear chromatin organization, cell proliferation, and transcription regulation were enriched. Similarly, pathways associated with aging, inflammatory response, innate immune response, and DNA damage were down-regulated. Our results highlight the role of L1 RNA in heterochromatin homeostasis in progeroid syndromes and identify a possible therapeutic approach to treat premature aging and related syndromes.


Assuntos
Senilidade Prematura , Síndrome de Cockayne , Proteínas Imediatamente Precoces , Progéria , Senilidade Prematura/genética , Animais , Antígenos de Diferenciação , Heterocromatina , Histonas/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Elementos Nucleotídeos Longos e Dispersos , Lisina/metabolismo , Camundongos , Fenótipo , Progéria/genética , RNA , Telômero/genética , Proteínas Supressoras de Tumor/genética
4.
Front Cell Dev Biol ; 10: 786031, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35309931

RESUMO

It is widely believed that cellular senescence plays a critical role in both aging and cancer, and that senescence is a fundamental, permanent growth arrest that somatic cells cannot avoid. Here we show that Myc plays an important role in self-renewal of esophageal epithelial cells, contributing to their resistance to cellular senescence. Myc is homogeneously expressed in basal cells of the esophageal epithelium and Myc positively regulates their self-renewal by maintaining their undifferentiated state. Indeed, Myc knockout induced a loss of the undifferentiated state of esophageal epithelial cells resulting in cellular senescence while forced MYC expression promoted oncogenic cell proliferation. A superoxide scavenger counteracted Myc knockout-induced senescence, therefore suggesting that a mitochondrial superoxide takes part in inducing senescence. Taken together, these analyses reveal extremely low levels of cellular senescence and senescence-associated phenotypes in the esophageal epithelium, as well as a critical role for Myc in self-renewal of basal cells in this organ. This provides new avenues for studying and understanding the links between stemness and resistance to cellular senescence.

5.
Nat Commun ; 12(1): 3094, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34035273

RESUMO

Short-term, systemic expression of the Yamanaka reprogramming factors (Oct-3/4, Sox2, Klf4 and c-Myc [OSKM]) has been shown to rejuvenate aging cells and promote tissue regeneration in vivo. However, the mechanisms by which OSKM promotes tissue regeneration are unknown. In this work, we focus on a specific tissue and demonstrate that local expression of OSKM, specifically in myofibers, induces the activation of muscle stem cells or satellite cells (SCs), which accelerates muscle regeneration in young mice. In contrast, expressing OSKM directly in SCs does not improve muscle regeneration. Mechanistically, expressing OSKM in myofibers regulates the expression of genes important for the SC microenvironment, including upregulation of p21, which in turn downregulates Wnt4. This is critical because Wnt4 is secreted by myofibers to maintain SC quiescence. Thus, short-term induction of the Yamanaka factors in myofibers may promote tissue regeneration by modifying the stem cell niche.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular/genética , Miofibrilas/metabolismo , Regeneração/genética , Células Satélites de Músculo Esquelético/metabolismo , Nicho de Células-Tronco , Animais , Células Cultivadas , Feminino , Expressão Gênica , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Camundongos Transgênicos , Miofibrilas/fisiologia , Fator 3 de Transcrição de Octâmero/genética , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição SOXB1/genética , Células Satélites de Músculo Esquelético/citologia , Proteína Wnt4/genética
6.
Cell ; 184(8): 2020-2032.e14, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33861963

RESUMO

Interspecies chimera formation with human pluripotent stem cells (hPSCs) represents a necessary alternative to evaluate hPSC pluripotency in vivo and might constitute a promising strategy for various regenerative medicine applications, including the generation of organs and tissues for transplantation. Studies using mouse and pig embryos suggest that hPSCs do not robustly contribute to chimera formation in species evolutionarily distant to humans. We studied the chimeric competency of human extended pluripotent stem cells (hEPSCs) in cynomolgus monkey (Macaca fascicularis) embryos cultured ex vivo. We demonstrate that hEPSCs survived, proliferated, and generated several peri- and early post-implantation cell lineages inside monkey embryos. We also uncovered signaling events underlying interspecific crosstalk that may help shape the unique developmental trajectories of human and monkey cells within chimeric embryos. These results may help to better understand early human development and primate evolution and develop strategies to improve human chimerism in evolutionarily distant species.


Assuntos
Quimerismo , Embrião de Mamíferos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Macaca fascicularis , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante , RNA-Seq , Análise de Célula Única , Transcriptoma
7.
Cell ; 179(3): 687-702.e18, 2019 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-31626770

RESUMO

A single mouse blastomere from an embryo until the 8-cell stage can generate an entire blastocyst. Whether laboratory-cultured cells retain a similar generative capacity remains unknown. Starting from a single stem cell type, extended pluripotent stem (EPS) cells, we established a 3D differentiation system that enabled the generation of blastocyst-like structures (EPS-blastoids) through lineage segregation and self-organization. EPS-blastoids resembled blastocysts in morphology and cell-lineage allocation and recapitulated key morphogenetic events during preimplantation and early postimplantation development in vitro. Upon transfer, some EPS-blastoids underwent implantation, induced decidualization, and generated live, albeit disorganized, tissues in utero. Single-cell and bulk RNA-sequencing analysis revealed that EPS-blastoids contained all three blastocyst cell lineages and shared transcriptional similarity with natural blastocysts. We also provide proof of concept that EPS-blastoids can be generated from adult cells via cellular reprogramming. EPS-blastoids provide a unique platform for studying early embryogenesis and pave the way to creating viable synthetic embryos by using cultured cells.


Assuntos
Blastocisto/citologia , Linhagem da Célula , Implantação do Embrião , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Embrionárias Murinas/citologia , Criação de Embriões para Pesquisa/métodos , Animais , Blastocisto/metabolismo , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Técnicas de Reprogramação Celular/métodos , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Células-Tronco Embrionárias Murinas/metabolismo , Transcriptoma
8.
Cell Res ; 29(10): 804-819, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31444470

RESUMO

In vivo genome editing represents a powerful strategy for both understanding basic biology and treating inherited diseases. However, it remains a challenge to develop universal and efficient in vivo genome-editing tools for tissues that comprise diverse cell types in either a dividing or non-dividing state. Here, we describe a versatile in vivo gene knock-in methodology that enables the targeting of a broad range of mutations and cell types through the insertion of a minigene at an intron of the target gene locus using an intracellularly linearized single homology arm donor. As a proof-of-concept, we focused on a mouse model of premature-aging caused by a dominant point mutation, which is difficult to repair using existing in vivo genome-editing tools. Systemic treatment using our new method ameliorated aging-associated phenotypes and extended animal lifespan, thus highlighting the potential of this methodology for a broad range of in vivo genome-editing applications.


Assuntos
Edição de Genes/métodos , Animais , Sistemas CRISPR-Cas/genética , Reparo do DNA , Dependovirus/genética , Fator de Transcrição GATA3/genética , Técnicas de Introdução de Genes , Terapia Genética/métodos , Vetores Genéticos/metabolismo , Células-Tronco Embrionárias Humanas , Humanos , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Neurônios/citologia , Neurônios/metabolismo , RNA Guia de Cinetoplastídeos/metabolismo , Ratos , Tubulina (Proteína)/genética
9.
Cell Stem Cell ; 25(3): 373-387.e9, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31303547

RESUMO

Human pluripotent stem cell-derived kidney organoids recapitulate developmental processes and tissue architecture, but intrinsic limitations, such as lack of vasculature and functionality, have greatly hampered their application. Here we establish a versatile protocol for generating vascularized three-dimensional (3D) kidney organoids. We employ dynamic modulation of WNT signaling to control the relative proportion of proximal versus distal nephron segments, producing a correlative level of vascular endothelial growth factor A (VEGFA) to define a resident vascular network. Single-cell RNA sequencing identifies a subset of nephron progenitor cells as a potential source of renal vasculature. These kidney organoids undergo further structural and functional maturation upon implantation. Using this kidney organoid platform, we establish an in vitro model of autosomal recessive polycystic kidney disease (ARPKD), the cystic phenotype of which can be effectively prevented by gene correction or drug treatment. Our studies provide new avenues for studying human kidney development, modeling disease pathogenesis, and performing patient-specific drug validation.


Assuntos
Rim/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Rim Policístico Autossômico Recessivo/patologia , Diferenciação Celular , Células Cultivadas , Descoberta de Drogas , Terapia Genética , Humanos , Rim/irrigação sanguínea , Neovascularização Fisiológica , Técnicas de Cultura de Órgãos , Organogênese , Organoides/irrigação sanguínea , Rim Policístico Autossômico Recessivo/metabolismo , Rim Policístico Autossômico Recessivo/terapia , Medicina de Precisão , Fator A de Crescimento do Endotélio Vascular/metabolismo , Via de Sinalização Wnt
10.
Protein Cell ; 10(7): 485-495, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31041783

RESUMO

Identification of the precise molecular pathways involved in oncogene-induced transformation may help us gain a better understanding of tumor initiation and promotion. Here, we demonstrate that SOX2+ foregut epithelial cells are prone to oncogenic transformation upon mutagenic insults, such as KrasG12D and p53 deletion. GFP-based lineage-tracing experiments indicate that SOX2+ cells are the cells-of-origin of esophagus and stomach hyperplasia. Our observations indicate distinct roles for oncogenic KRAS mutation and P53 deletion. p53 homozygous deletion is required for the acquisition of an invasive potential, and KrasG12D expression, but not p53 deletion, suffices for tumor formation. Global gene expression analysis reveals secreting factors upregulated in the hyperplasia induced by oncogenic KRAS and highlights a crucial role for the CXCR2 pathway in driving hyperplasia. Collectively, the array of genetic models presented here demonstrate that stratified epithelial cells are susceptible to oncogenic insults, which may lead to a better understanding of tumor initiation and aid in the design of new cancer therapeutics.


Assuntos
Neoplasias Esofágicas/metabolismo , Mutação , Receptores de Interleucina-8B/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Proliferação de Células , Neoplasias Esofágicas/patologia , Feminino , Masculino , Camundongos , Camundongos Mutantes , Transdução de Sinais , Células Tumorais Cultivadas
11.
Cell ; 167(7): 1719-1733.e12, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984723

RESUMO

Aging is the major risk factor for many human diseases. In vitro studies have demonstrated that cellular reprogramming to pluripotency reverses cellular age, but alteration of the aging process through reprogramming has not been directly demonstrated in vivo. Here, we report that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in a mouse model of premature aging. Similarly, expression of OSKM in vivo improves recovery from metabolic disease and muscle injury in older wild-type mice. The amelioration of age-associated phenotypes by epigenetic remodeling during cellular reprogramming highlights the role of epigenetic dysregulation as a driver of mammalian aging. Establishing in vivo platforms to modulate age-associated epigenetic marks may provide further insights into the biology of aging.


Assuntos
Envelhecimento/genética , Reprogramação Celular , Epigênese Genética , Doenças Metabólicas/genética , Fatores de Transcrição/metabolismo , Senilidade Prematura/genética , Senilidade Prematura/metabolismo , Animais , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Lamina Tipo A/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/prevenção & controle , Camundongos , Modelos Animais , Pâncreas/metabolismo , Sarcopenia/metabolismo
12.
Cell Stem Cell ; 19(4): 516-529, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27570066

RESUMO

Transit-amplifying nephron progenitor cells (NPCs) generate all of the nephrons of the mammalian kidney during development. Their limited numbers, poor in vitro expansion, and difficult accessibility in humans have slowed basic and translational research into renal development and diseases. Here, we show that with appropriate 3D culture conditions, it is possible to support long-term expansion of primary mouse and human fetal NPCs as well as NPCs derived from human induced pluripotent stem cells (iPSCs). Expanded NPCs maintain genomic stability, molecular homogeneity, and nephrogenic potential in vitro, ex vivo, and in vivo. Cultured NPCs are amenable to gene targeting and can form nephron organoids that engraft in vivo, functionally couple to the host's circulatory system, and produce urine-like metabolites via filtration. Together, these findings provide a technological platform for studying human nephrogenesis, modeling and diagnosing renal diseases, and drug discovery.


Assuntos
Técnicas de Cultura de Células/métodos , Néfrons/citologia , Organogênese , Células-Tronco/citologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Edição de Genes , Humanos , Testes de Função Renal , Camundongos , Organoides/citologia , Comunicação Parácrina , Células-Tronco/metabolismo , Fatores de Tempo
13.
Cell Stem Cell ; 15(5): 589-604, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25517466

RESUMO

Heart failure is a leading cause of mortality and morbidity in the developed world, partly because mammals lack the ability to regenerate heart tissue. Whether this is due to evolutionary loss of regenerative mechanisms present in other organisms or to an inability to activate such mechanisms is currently unclear. Here we decipher mechanisms underlying heart regeneration in adult zebrafish and show that the molecular regulators of this response are conserved in mammals. We identified miR-99/100 and Let-7a/c and their protein targets smarca5 and fntb as critical regulators of cardiomyocyte dedifferentiation and heart regeneration in zebrafish. Although human and murine adult cardiomyocytes fail to elicit an endogenous regenerative response after myocardial infarction, we show that in vivo manipulation of this molecular machinery in mice results in cardiomyocyte dedifferentiation and improved heart functionality after injury. These data provide a proof of concept for identifying and activating conserved molecular programs to regenerate the damaged heart.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/fisiologia , Mamíferos/genética , MicroRNAs/genética , Regeneração/genética , Animais , Desdiferenciação Celular/genética , Proliferação de Células , Regulação para Baixo/genética , Inativação Gênica , Genoma , Humanos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Peixe-Zebra/genética
14.
Nat Protoc ; 9(11): 2693-704, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25340442

RESUMO

This protocol presents recently developed methodologies for the differentiation of human pluripotent stem cells (hPSCs) into ureteric bud (UB) progenitor-like cells. Differentiation of human PSCs to UB progenitor-like cells allows for the generation of chimeric kidney cultures in which the human cells can self-assemble into chimeric 3D structures in combination with embryonic mouse kidney cells over a period of 18 d. UB progenitor-like cells are generated by a two-step process that combines in vitro commitment of human PSCs, whether embryonic stem cells (ESCs) or induced PSCs (iPSCs), under chemically defined culture conditions, with ex vivo cultures for the induction of 3D organogenesis. The models described here provide new opportunities for investigating human kidney development, modeling disease, evaluating regenerative medicine strategies, as well as for toxicology studies.


Assuntos
Técnicas de Cultura de Células , Rim/citologia , Técnicas de Cultura de Órgãos/métodos , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Feminino , Humanos , Rim/embriologia , Masculino , Camundongos Endogâmicos , Organogênese
15.
J Biol Chem ; 289(3): 1788-97, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24311780

RESUMO

Nodal, a member of the TGF-ß superfamily, plays an important role in vertebrate and invertebrate early development. The biochemical study of Nodal and its signaling pathway has been a challenge, mainly because of difficulties in producing the protein in sufficient quantities. We have developed a library of stable, chemically refoldable Nodal/BMP2 chimeric ligands (NB2 library). Three chimeras, named NB250, NB260, and NB264, show Nodal-like signaling properties including dependence on the co-receptor Cripto and activation of the Smad2 pathway. NB250, like Nodal, alters heart looping during the establishment of embryonic left-right asymmetry, and both NB250 and NB260, as well as Nodal, induce chondrogenic differentiation of human adipose-derived stem cells. This Nodal-induced differentiation is shown to be more efficient than BPM2-induced differentiation. Interestingly, the crystal structure of NB250 shows a backbone scaffold similar to that of BMP2. Our results show that these chimeric ligands may have therapeutic implications in cartilage injuries.


Assuntos
Tecido Adiposo/metabolismo , Proteína Morfogenética Óssea 2 , Condrogênese/efeitos dos fármacos , Proteína Nodal , Proteínas Recombinantes de Fusão , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/metabolismo , Tecido Adiposo/patologia , Adulto , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/farmacologia , Cartilagem/lesões , Cartilagem/metabolismo , Cartilagem/patologia , Linhagem Celular , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Nodal/química , Proteína Nodal/genética , Proteína Nodal/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Células-Tronco/patologia
16.
Dev Biol ; 327(1): 177-90, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19133254

RESUMO

Epimorphic regeneration is a unique and complex instance of postembryonic growth observed in certain metazoans that is usually triggered by severe injury [Akimenko et al., 2003; Alvarado and Tsonis, 2006; Brockes, 1997; Endo et al., 2004]. Cell division and migration are two fundamental biological processes required for supplying replacement cells during regeneration [Endo et al., 2004; Slack, 2007]. However, the connection between the early stimuli generated after injury and the signals regulating proliferation and migration during regeneration remain largely unknown. Here we show that the oncogenes ErbB2 and ErbB3, two members of the EGFR family, are essential for mounting a successful regeneration response in vertebrates. Importantly, amputation-induced progenitor proliferation and migration are significantly reduced upon genetic and/or chemical modulation of ErbB function. Moreover, we also found that NRG1 and PI3K functionally interact with ErbB2 and ErbB3 during regeneration and interfering with their function also abrogates the capacity of progenitor cells to regenerate lost structures upon amputation. Our findings suggest that ErbB, PI3K and NRG1 are components of a permissive switch for migration and proliferation continuously acting across the amputated fin from early stages of vertebrate regeneration onwards that regulate the expression of the transcription factors lef1 and msxB.


Assuntos
Amputação Cirúrgica , Receptor ErbB-2/fisiologia , Receptor ErbB-3/fisiologia , Regeneração , Células-Tronco/fisiologia , Animais , Movimento Celular , Proliferação de Células , Proteínas de Homeodomínio/genética , Neuregulina-1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/genética , Vertebrados , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
17.
Development ; 136(4): 585-94, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19168674

RESUMO

The genetic mechanisms that regulate the complex morphogenesis of generating cartilage elements in correct positions with precise shapes during organogenesis, fundamental issues in developmental biology, are still not well understood. By focusing on the developing mouse limb, we confirm the importance of transcription factors encoded by the Sall gene family in proper limb morphogenesis, and further show that they have overlapping activities in regulating regional morphogenesis in the autopod. Sall1/Sall3 double null mutants exhibit a loss of digit1 as well as a loss or fusion of digit2 and digit3, metacarpals and carpals in the autopod. We show that Sall activity affects different pathways, including the Shh signaling pathway, as well as the Hox network. Shh signaling in the mesenchyme is partially impaired in the Sall mutant limbs. Additionally, our data suggest an antagonism between Sall1-Sall3 and Hoxa13-Hoxd13. We demonstrate that expression of Epha3 and Epha4 is downregulated in the Sall1/Sall3 double null mutants, and, conversely, is upregulated in Hoxa13 and Hoxd13 mutants. Moreover, the expression of Sall1 and Sall3 is upregulated in Hoxa13 and Hoxd13 mutants. Furthermore, by using DNA-binding assays, we show that Sall and Hox compete for a target sequence in the Epha4 upstream region. In conjunction with the Shh pathway, the antagonistic interaction between Hoxa13-Hoxd13 and Sall1-Sall3 in the developing limb may contribute to the fine-tuning of local Hox activity that leads to proper morphogenesis of each cartilage element of the vertebrate autopod.


Assuntos
Extremidades/embriologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Morfogênese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Especificidade de Órgãos , Receptor EphA3/metabolismo , Receptor EphA4/metabolismo , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Proteína Gli3 com Dedos de Zinco
18.
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
19.
PLoS One ; 2(4): e398, 2007 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-17460765

RESUMO

BACKGROUND: The heart forms from a linear tube that is subject to complex remodeling during embryonic development. Hallmarks of this remodeling are the looping of the heart tube and the regionalization into chamber and non-chamber myocardium. Cardiomyocytes in the future chamber myocardium acquire different cellular and physiological characteristics through activation of a chamber-specific genetic program, which is in part mediated by T-box genes. METHODOLOGY/PRINCIPAL FINDING: We characterize two new zebrafish T-box transcription factors, tbx3b and tbx2a, and analyze their role during the development of the atrioventricular canal. Loss- and gain-of-function analyses demonstrate that tbx3b and tbx2a are necessary to repress the chamber-genetic program in the non-chamber myocardium. We also show that tbx3b and tbx2a are required to control cell proliferation in the atrioventricular canal and that misregulation of cell proliferation in the heart tube influences looping. Furthermore, we characterize the heart phenotype of a novel Tbx3 mutation in mice and show that both the control of cell proliferation and the repression of chamber-specific genetic program in the non-chamber myocardium are conserved roles of Tbx3 in this species. CONCLUSIONS/SIGNIFICANCE: Taken together, our results uncover an evolutionarily conserved role of Tbx2/3 transcription factors during remodeling of the heart myocardium and highlight the importance of controlling cell proliferation as a driving force of morphogenesis.


Assuntos
Proliferação de Células , Coração/fisiologia , Fatores de Transcrição/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/genética , Peixe-Zebra
20.
Nat Clin Pract Cardiovasc Med ; 4 Suppl 1: S77-82, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17230219

RESUMO

The migration of myocardial precursor cells towards the embryonic midline underlies the formation of the heart tube and is a key process of heart organogenesis. The zebrafish mutation miles-apart (mil), which affects the gene encoding a sphingosine-1-phosphate receptor, is characterized by defective migration of myocardial precursor cells and results in the formation of two laterally positioned hearts, a condition known as cardia bifida. The mechanism that disrupts myocardial migration in mil mutants remains largely unclear. To investigate how mil regulates this process, here we analyze the interactions between mil and other mediators of myocardial migration. We show that mil function is associated with the other known cardia bifida locus, natter/fibronectin (nat/fn), which encodes fibronectin, a major component of the extracellular matrix, in the control of myocardial migration. By using a primary culture system of embryonic zebrafish cells, we also show that signaling from the sphingosine-1-phosphate receptor regulates cell-fibronectin interactions in zebrafish. In addition, localized inhibition and activation of cell-fibronectin interactions during the stages of myocardial migration reveal that the temporal regulation of cell-fibronectin interaction by mil is required for proper myocardial migration. Our study reveals novel functional links between sphingosine-1-phosphate receptor signaling and cell-fibronectin interaction in the control of myocardial migration during zebrafish heart organogenesis.


Assuntos
Fibronectinas/fisiologia , Miócitos Cardíacos/fisiologia , Organogênese/fisiologia , Receptores de Lisoesfingolipídeo/genética , Animais , Comunicação Celular , Movimento Celular , Genótipo , Mutação , Organogênese/genética , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...