Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 535(7612): 430-4, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27398620

RESUMO

Insulin-dependent diabetes is a complex multifactorial disorder characterized by loss or dysfunction of ß-cells. Pancreatic ß-cells differ in size, glucose responsiveness, insulin secretion and precursor cell potential; understanding the mechanisms that underlie this functional heterogeneity might make it possible to develop new regenerative approaches. Here we show that Fltp (also known as Flattop and Cfap126), a Wnt/planar cell polarity (PCP) effector and reporter gene acts as a marker gene that subdivides endocrine cells into two subpopulations and distinguishes proliferation-competent from mature ß-cells with distinct molecular, physiological and ultrastructural features. Genetic lineage tracing revealed that endocrine subpopulations from Fltp-negative and -positive lineages react differently to physiological and pathological changes. The expression of Fltp increases when endocrine cells cluster together to form polarized and mature 3D islet mini-organs. We show that 3D architecture and Wnt/PCP ligands are sufficient to trigger ß-cell maturation. By contrast, the Wnt/PCP effector Fltp is not necessary for ß-cell development, proliferation or maturation. We conclude that 3D architecture and Wnt/PCP signalling underlie functional ß-cell heterogeneity and induce ß-cell maturation. The identification of Fltp as a marker for endocrine subpopulations sheds light on the molecular underpinnings of islet cell heterogeneity and plasticity and might enable targeting of endocrine subpopulations for the regeneration of functional ß-cell mass in diabetic patients.


Assuntos
Ilhotas Pancreáticas/citologia , Animais , Biomarcadores/análise , Diferenciação Celular , Linhagem da Célula/genética , Polaridade Celular , Proliferação de Células , Humanos , Resistência à Insulina , Ilhotas Pancreáticas/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Via de Sinalização Wnt
2.
Proc Natl Acad Sci U S A ; 109(19): 7356-61, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22529374

RESUMO

During early pancreatic development, Notch signaling represses differentiation of endocrine cells and promotes proliferation of Nkx6-1(+)Ptf1a(+) multipotent progenitor cells (MPCs). Later, antagonistic interactions between Nkx6 transcription factors and Ptf1a function to segregate MPCs into distal Nkx6-1(-)Ptf1a(+) acinar progenitors and proximal Nkx6-1(+)Ptf1a(-) duct and ß-cell progenitors. Distal cells are initially multipotent, but evolve into unipotent, acinar cell progenitors. Conversely, proximal cells are bipotent and give rise to duct cells and late-born endocrine cells, including the insulin producing ß-cells. However, signals that regulate proximodistal (P-D) patterning and thus formation of ß-cell progenitors are unknown. Here we show that Mind bomb 1 (Mib1) is required for correct P-D patterning of the developing pancreas and ß-cell formation. We found that endoderm-specific inactivation of Mib1 caused a loss of Nkx6-1(+)Ptf1a(-) and Hnf1ß(+) cells and a corresponding loss of Neurog3(+) endocrine progenitors and ß-cells. An accompanying increase in Nkx6-1(-)Ptf1a(+) and amylase(+) cells, occupying the proximal domain, suggests that proximal cells adopt a distal fate in the absence of Mib1 activity. Impeding Notch-mediated transcriptional activation by conditional expression of dominant negative Mastermind-like 1 (Maml1) resulted in a similarly distorted P-D patterning and suppressed ß-cell formation, as did conditional inactivation of the Notch target gene Hes1. Our results reveal iterative use of Notch in pancreatic development to ensure correct P-D patterning and adequate ß-cell formation.


Assuntos
Embrião de Mamíferos/metabolismo , Células Secretoras de Insulina/metabolismo , Pâncreas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Pâncreas/citologia , Pâncreas/embriologia , Receptores Notch/genética , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/genética
3.
Differentiation ; 83(2): S105-13, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22153975

RESUMO

We recently identified Flattop (Fltp; 1700009p17Rik) in a screen for potential Foxa2 target and novel mouse organizer genes. Besides its expression in the embryonic node, we found that Fltp is active in other monociliated tissues such as the sensory organs of the inner ear, duct and islets of the pancreas as well as in testis. Additionally, Fltp mRNA is expressed in multiciliated epithelial cells of the lung and of the choroid plexi in the brain. To genetically lineage trace these cells during development and injury as well as to conditionally inactivate genes in these tissues, we generated a Cre recombinase knock-in mouse line using the Fltp gene locus. By homologous recombination we have fused the Fltp open-reading frame to a tandem affinity purification (TAP) tag followed by an intervening viral T2A sequence for co-translational cleavage and an improved Cre recombinase (iCre). This strategy allows both the analysis of the tagged Fltp-TAP-T2A protein and the usage of the iCre recombinase for conditional targeting approaches. Using the ROSA26 reporter mouse line we show that Fltp(T2AiCre) is first active in the monociliated cells of the node, notochord, floorplate and prechordal plate, consistent with the Fltp-TAP-T2A protein production in the node progenitor cells. Furthermore iCre recombinase activity is detected in multiciliated tissues such as choroid plexi of the brain and epithelial cells of the lung with the onset at E10.5 and E13.5, respectively. In the pancreas, ß-galactosidase activity is seen in the monociliated cells of the pancreatic duct and islet of Langerhans. Intercrossing Fltp(T2AiCre) mice with the CAG-CAT-EGFP reporter mouse line further confirms iCre activity in multiciliated cells of the lung and brain on a cellular level. Thus, the Fltp(T2AiCre) line is a powerful tool to conditionally inactivate genes in distinct mono- and multiciliated tissues and to analyze the tagged Fltp protein in vivo.


Assuntos
Técnicas de Introdução de Genes , Marcação de Genes , Modelos Animais , Animais , Linhagem Celular , Cílios/metabolismo , Células-Tronco Embrionárias/metabolismo , Genótipo , Imuno-Histoquímica , Integrases/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Pâncreas/metabolismo
4.
Diabetes ; 66(5): 1247-1257, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28174288

RESUMO

In pancreatic ß-cells, mitochondrial bioenergetics control glucose-stimulated insulin secretion. Mitochondrial dynamics are generally associated with quality control, maintaining the functionality of bioenergetics. By acute pharmacological inhibition of mitochondrial fission protein Drp1, we demonstrate in this study that mitochondrial fission is necessary for glucose-stimulated insulin secretion in mouse and human islets. We confirm that genetic silencing of Drp1 increases mitochondrial proton leak in MIN6 cells. However, our comprehensive analysis of pancreatic islet bioenergetics reveals that Drp1 does not control insulin secretion via its effect on proton leak but instead via modulation of glucose-fueled respiration. Notably, pyruvate fully rescues the impaired insulin secretion of fission-deficient ß-cells, demonstrating that defective mitochondrial dynamics solely affect substrate supply upstream of oxidative phosphorylation. The present findings provide novel insights into how mitochondrial dysfunction may cause pancreatic ß-cell failure. In addition, the results will stimulate new thinking in the intersecting fields of mitochondrial dynamics and bioenergetics, as treatment of defective dynamics in mitochondrial diseases appears to be possible by improving metabolism upstream of mitochondria.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/genética , Trifosfato de Adenosina/metabolismo , Animais , Dinaminas/antagonistas & inibidores , Metabolismo Energético/genética , GTP Fosfo-Hidrolases/antagonistas & inibidores , GTP Fosfo-Hidrolases/genética , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Humanos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/patologia , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Ácido Pirúvico/farmacologia
5.
Nat Rev Endocrinol ; 12(12): 695-709, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27585958

RESUMO

Although ß-cell heterogeneity was discovered more than 50 years ago, the underlying principles have been explored only during the past decade. Islet-cell heterogeneity arises during pancreatic development and might reflect the existence of distinct populations of progenitor cells and the developmental pathways of endocrine cells. Heterogeneity can also be acquired in the postnatal period owing to ß-cell plasticity or changes in islet architecture. Furthermore, ß-cell neogenesis, replication and dedifferentiation represent alternative sources of ß-cell heterogeneity. In addition to a physiological role, ß-cell heterogeneity influences the development of diabetes mellitus and its response to treatment. Identifying phenotypic and functional markers to discriminate distinct ß-cell subpopulations and the mechanisms underpinning their regulation is warranted to advance current knowledge of ß-cell function and to design novel regenerative strategies that target subpopulations of ß cells. In this context, the Wnt/planar cell polarity (PCP) effector molecule Flattop can distinguish two unique ß-cell subpopulations with specific transcriptional signatures, functional properties and differential responses to environmental stimuli. In vivo targeting of these ß-cell subpopulations might, therefore, represent an alternative strategy for the future treatment of diabetes mellitus.


Assuntos
Diferenciação Celular , Linhagem da Célula , Plasticidade Celular , Polaridade Celular , Células Secretoras de Insulina/citologia , Células-Tronco/citologia , Animais , Humanos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Proteínas Associadas aos Microtúbulos , Transcriptoma
6.
Elife ; 32014 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25296022

RESUMO

Planar cell polarity (PCP) regulates basal body (BB) docking and positioning during cilia formation, but the underlying mechanisms remain elusive. In this study, we investigate the uncharacterized gene Flattop (Fltp) that is transcriptionally activated during PCP acquisition in ciliated tissues. Fltp knock-out mice show BB docking and ciliogenesis defects in multiciliated lung cells. Furthermore, Fltp is necessary for kinocilium positioning in monociliated inner ear hair cells. In these cells, the core PCP molecule Dishevelled 2, the BB/spindle positioning protein Dlg3, and Fltp localize directly adjacent to the apical plasma membrane, physically interact and surround the BB at the interface of the microtubule and actin cytoskeleton. Dlg3 and Fltp knock-outs suggest that both cooperatively translate PCP cues for BB positioning in the inner ear. Taken together, the identification of novel BB/spindle positioning components as potential mediators of PCP signaling might have broader implications for other cell types, ciliary disease, and asymmetric cell division.


Assuntos
Corpos Basais/metabolismo , Cílios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Corpos Basais/ultraestrutura , Sítios de Ligação , Polaridade Celular , Cílios/ultraestrutura , Sequência Conservada , Orelha Interna/metabolismo , Orelha Interna/ultraestrutura , Genes Reporter , Fator 3-beta Nuclear de Hepatócito/metabolismo , Pulmão/patologia , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/genética , Dados de Sequência Molecular , Morfogênese , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas/química , Proteínas/genética , Transdução de Sinais , Estereocílios/metabolismo , Estereocílios/ultraestrutura , Junções Íntimas/metabolismo
8.
Cardiovasc Res ; 92(1): 115-22, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21632880

RESUMO

AIMS: Our recent work demonstrated that common cardiovascular progenitor cells are characterized and induced by the expression of the transcription factor mesoderm posterior1 (MesP1) in vertebrate embryos and murine embryonic stem cells. As the proliferative potential of stem cell-derived cardiomyocytes is limited, it is crucial to understand how MesP1 expression is mediated in order to achieve reasonable and reliable yields for novel stem cell-based therapeutic options. As potential upstream regulators of MesP1, we therefore analysed Eomes and Brachyury(T), which had been controversially discussed as being crucial for cardiovasculogenic lineage formation. METHODS AND RESULTS: Wild-type and transgenic murine embryonic stem cell lines, mRNA analyses, embryoid body formation, and cell sorting revealed that the MesP1 positive population emerges from the Brachyury(T) positive fraction. In situ hybridizations using wild-type mouse embryos confirmed that Brachyury(T) colocalises with MesP1 in vivo. Likewise, shRNA-based loss of Brachyury(T) causes a dramatic decrease in MesP1 expression accompanied by reduced cardiac markers in differentiating embryonic stem cells, which is reflected in vivo via in situ hybridizations using Brachyury(T) knock-out embryos where MesP1 mRNA is greatly abolished. We finally defined a 3.4 kb proximal MesP1-promoter fragment which is directly bound and activated by Brachyury(T) via a T responsive element as shown via bandshift, chromatin immuneprecipitation, and reporter assays. CONCLUSION: Our work contributes to the understanding of the earliest cardiovasculogenic events and may become an important prerequisite for cell therapy, tissue engineering, and pharmacological testing in the culture dish using pluripotent stem cell-derived as well as directly reprogrammed cardiovascular cell types.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Sistema Cardiovascular/citologia , Proteínas Fetais/fisiologia , Células-Tronco Multipotentes/fisiologia , Miócitos Cardíacos/citologia , Proteínas com Domínio T/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Células-Tronco Embrionárias/citologia , Humanos , Camundongos
9.
Dev Cell ; 21(3): 479-91, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21920314

RESUMO

The Drosophila Discs large (Dlg) scaffolding protein acts as a tumor suppressor regulating basolateral epithelial polarity and proliferation. In mammals, four Dlg homologs have been identified; however, their functions in cell polarity remain poorly understood. Here, we demonstrate that the X-linked mental retardation gene product Dlg3 contributes to apical-basal polarity and epithelial junction formation in mouse organizer tissues, as well as to planar cell polarity in the inner ear. We purified complexes associated with Dlg3 in polarized epithelial cells, including proteins regulating directed trafficking and tight junction formation. Remarkably, of the four Dlg family members, Dlg3 exerts a distinct function by recruiting the ubiquitin ligases Nedd4 and Nedd4-2 through its PPxY motifs. We found that these interactions are required for Dlg3 monoubiquitination, apical membrane recruitment, and tight junction consolidation. Our findings reveal an unexpected evolutionary diversification of the vertebrate Dlg family in basolateral epithelium formation.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Junções Íntimas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Animais , Polaridade Celular/fisiologia , Orelha Interna/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Ubiquitina-Proteína Ligases Nedd4 , Transporte Proteico , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA