Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 137(16): 2139-2151, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33667305

RESUMO

We studied a subset of hematopoietic stem cells (HSCs) that are defined by elevated expression of CD41 (CD41hi) and showed bias for differentiation toward megakaryocytes (Mks). Mouse models of myeloproliferative neoplasms (MPNs) expressing JAK2-V617F (VF) displayed increased frequencies and percentages of the CD41hi vs CD41lo HSCs compared with wild-type controls. An increase in CD41hi HSCs that correlated with JAK2-V617F mutant allele burden was also found in bone marrow from patients with MPN. CD41hi HSCs produced a higher number of Mk-colonies of HSCs in single-cell cultures in vitro, but showed reduced long-term reconstitution potential compared with CD41lo HSCs in competitive transplantations in vivo. RNA expression profiling showed an upregulated cell cycle, Myc, and oxidative phosphorylation gene signatures in CD41hi HSCs, whereas CD41lo HSCs showed higher gene expression of interferon and the JAK/STAT and TNFα/NFκB signaling pathways. Higher cell cycle activity and elevated levels of reactive oxygen species were confirmed in CD41hi HSCs by flow cytometry. Expression of Epcr, a marker for quiescent HSCs inversely correlated with expression of CD41 in mice, but did not show such reciprocal expression pattern in patients with MPN. Treatment with interferon-α further increased the frequency and percentage of CD41hi HSCs and reduced the number of JAK2-V617F+ HSCs in mice and patients with MPN. The shift toward the CD41hi subset of HSCs by interferon-α provides a possible mechanism of how interferon-α preferentially targets the JAK2 mutant clone.


Assuntos
Interferon-alfa/uso terapêutico , Janus Quinase 2/genética , Megacariócitos/metabolismo , Transtornos Mieloproliferativos/genética , Animais , Técnicas de Introdução de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Megacariócitos/citologia , Camundongos , Camundongos Transgênicos , Transtornos Mieloproliferativos/tratamento farmacológico , Glicoproteína IIb da Membrana de Plaquetas/genética , Mutação Puntual/efeitos dos fármacos
2.
Nature ; 573(7775): E5, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31515536

RESUMO

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nature ; 573(7774): 426-429, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31485073

RESUMO

Haematopoietic stem cells self-renew and differentiate into all blood lineages throughout life, and can repair damaged blood systems upon transplantation. Asymmetric cell division has previously been suspected to be a regulator of haematopoietic-stem-cell fate, but its existence has not directly been shown1. In asymmetric cell division, asymmetric fates of future daughter cells are prospectively determined by a mechanism that is linked to mitosis. This can be mediated by asymmetric inheritance of cell-extrinsic niche signals by, for example, orienting the divisional plane, or by the asymmetric inheritance of cell-intrinsic fate determinants. Observations of asymmetric inheritance or of asymmetric daughter-cell fates alone are not sufficient to demonstrate asymmetric cell division2. In both cases, sister-cell fates could be controlled by mechanisms that are independent of division. Here we demonstrate that the cellular degradative machinery-including lysosomes, autophagosomes, mitophagosomes and the protein NUMB-can be asymmetrically inherited into haematopoietic-stem-cell daughter cells. This asymmetric inheritance predicts the asymmetric future metabolic and translational activation and fates of haematopoietic-stem-cell daughter cells and their offspring. Therefore, our studies provide evidence for the existence of asymmetric cell division in haematopoietic stem cells.

4.
Bioinformatics ; 35(13): 2291-2299, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30452534

RESUMO

MOTIVATION: Recent imaging technologies allow for high-throughput tracking of cells as they migrate, divide, express fluorescent markers and change their morphology. The interpretation of these data requires unbiased, efficient statistical methods that model the dynamics of cell phenotypes. RESULTS: We introduce treeHFM, a probabilistic model which generalizes the theory of hidden Markov models to tree structured data. While accounting for the entire genealogy of a cell, treeHFM categorizes cells according to their primary phenotypic features. It models all relevant events in a cell's life, including cell division, and thereby enables the analysis of event order and cell fate heterogeneity. Simulations show higher accuracy in predicting correct state labels when modeling the more complex, tree-shaped dependency of samples over standard HMM modeling. Applying treeHFM to time lapse images of hematopoietic progenitor cell differentiation, we demonstrate that progenitor cells undergo a well-ordered sequence of differentiation events. AVAILABILITY AND IMPLEMENTATION: The treeHFM is implemented in C++. We provide wrapper functions for the programming languages R (CRAN package, https://CRAN.R-project.org/package=treeHFM) and Matlab (available at Mathworks Central, http://se.mathworks.com/matlabcentral/fileexchange/57575-treehfml). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Imagem com Lapso de Tempo , Análise por Conglomerados , Modelos Estatísticos , Linguagens de Programação , Software
5.
Blood ; 129(12): 1691-1701, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28159742

RESUMO

Controlled regulation of lineage decisions is imperative for hematopoiesis. Yet, the molecular mechanisms underlying hematopoietic lineage choices are poorly defined. Colony-stimulating factor 1 (CSF-1), the cytokine acting as the principal regulator of monocyte/macrophage (M) development, has been shown to be able to instruct the lineage choice of uncommitted granulocyte M (GM) progenitors toward an M fate. However, the intracellular signaling pathways involved are unknown. CSF-1 activates a multitude of signaling pathways resulting in a pleiotropic cellular response. The precise role of individual pathways within this complex and redundant signaling network is dependent on cellular context, and is not well understood. Here, we address which CSF-1-activated pathways are involved in transmitting the lineage-instructive signal in primary bone marrow-derived GM progenitors. Although its loss is compensated for by alternative signaling activation mechanisms, Src family kinase (SFK) signaling is sufficient to transmit the CSF-1 lineage instructive signal. Moreover, c-Src activity is sufficient to drive M fate, even in nonmyeloid cells.


Assuntos
Linhagem da Célula , Fator Estimulador de Colônias de Macrófagos/fisiologia , Monócitos/citologia , Transdução de Sinais , Quinases da Família src/metabolismo , Animais , Células Cultivadas , Células Precursoras de Granulócitos/citologia , Hematopoese , Camundongos
6.
Nat Methods ; 14(4): 403-406, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28218899

RESUMO

Differentiation alters molecular properties of stem and progenitor cells, leading to changes in their shape and movement characteristics. We present a deep neural network that prospectively predicts lineage choice in differentiating primary hematopoietic progenitors using image patches from brightfield microscopy and cellular movement. Surprisingly, lineage choice can be detected up to three generations before conventional molecular markers are observable. Our approach allows identification of cells with differentially expressed lineage-specifying genes without molecular labeling.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Processamento de Imagem Assistida por Computador/métodos , Redes Neurais de Computação , Imagem com Lapso de Tempo/métodos , Animais , Área Sob a Curva , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Técnicas de Introdução de Genes , Aprendizado de Máquina , Masculino , Camundongos Mutantes , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/genética , Transativadores/metabolismo
7.
Sci Rep ; 6: 37462, 2016 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-27892478

RESUMO

Functionally distinct plasmacytoid and conventional dendritic cells (pDC and cDC) shape innate and adaptive immunity. They are derived from common dendritic cell progenitors (CDPs) in the murine bone marrow, which give rise to CD11c+ MHCII- precursors with early commitment to DC subpopulations. In this study, we dissect pDC development from CDP into an ordered sequence of differentiation events by monitoring the expression of CD11c, MHC class II, Siglec H and CCR9 in CDP cultures by continuous single cell imaging and tracking. Analysis of CDP genealogies revealed a stepwise differentiation of CDPs into pDCs in a part of the CDP colonies. This developmental pathway involved an early CD11c+ SiglecH- pre-DC stage and a Siglec H+ CCR9low precursor stage, which was followed rapidly by upregulation of CCR9 indicating final pDC differentiation. In the majority of the remaining CDP pedigrees however the Siglec H+ CCR9low precursor state was maintained for several generations. Thus, although a fraction of CDPs transits through precursor stages rapidly to give rise to a first wave of pDCs, the majority of CDP progeny differentiate more slowly and give rise to longer lived precursor cells which are poised to differentiate on demand.


Assuntos
Células da Medula Óssea/citologia , Linhagem da Célula/imunologia , Células Dendríticas/citologia , Análise de Célula Única/métodos , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Antígeno CD11c/genética , Antígeno CD11c/imunologia , Diferenciação Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fêmur/citologia , Fêmur/imunologia , Fêmur/metabolismo , Citometria de Fluxo , Expressão Gênica , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Receptores CCR/genética , Receptores CCR/imunologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/genética , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Tíbia/citologia , Tíbia/imunologia , Tíbia/metabolismo
9.
Blood ; 128(9): 1181-92, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27365423

RESUMO

The maintenance of hematopoietic stem cells (HSCs) during ex vivo culture is an important prerequisite for their therapeutic manipulation. However, despite intense research, culture conditions for robust maintenance of HSCs are still missing. Cultured HSCs are quickly lost, preventing their improved analysis and manipulation. Identification of novel factors supporting HSC ex vivo maintenance is therefore necessary. Coculture with the AFT024 stroma cell line is capable of maintaining HSCs ex vivo long-term, but the responsible molecular players remain unknown. Here, we use continuous long-term single-cell observation to identify the HSC behavioral signature under supportive or nonsupportive stroma cocultures. We report early HSC survival as a major characteristic of HSC-maintaining conditions. Behavioral screening after manipulation of candidate molecules revealed that the extracellular matrix protein dermatopontin (Dpt) is involved in HSC maintenance. DPT knockdown in supportive stroma impaired HSC survival, whereas ectopic expression of the Dpt gene or protein in nonsupportive conditions restored HSC survival. Supplementing defined stroma- and serum-free culture conditions with recombinant DPT protein improved HSC clonogenicity. These findings illustrate a previously uncharacterized role of Dpt in maintaining HSCs ex vivo.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Proteoglicanas de Sulfatos de Condroitina/genética , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/farmacologia , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Transgênicos , Células Estromais/citologia , Células Estromais/metabolismo , Fatores de Tempo
10.
Nature ; 535(7611): 299-302, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27411635

RESUMO

The mechanisms underlying haematopoietic lineage decisions remain disputed. Lineage-affiliated transcription factors with the capacity for lineage reprogramming, positive auto-regulation and mutual inhibition have been described as being expressed in uncommitted cell populations. This led to the assumption that lineage choice is cell-intrinsically initiated and determined by stochastic switches of randomly fluctuating cross-antagonistic transcription factors. However, this hypothesis was developed on the basis of RNA expression data from snapshot and/or population-averaged analyses. Alternative models of lineage choice therefore cannot be excluded. Here we use novel reporter mouse lines and live imaging for continuous single-cell long-term quantification of the transcription factors GATA1 and PU.1 (also known as SPI1). We analyse individual haematopoietic stem cells throughout differentiation into megakaryocytic-erythroid and granulocytic-monocytic lineages. The observed expression dynamics are incompatible with the assumption that stochastic switching between PU.1 and GATA1 precedes and initiates megakaryocytic-erythroid versus granulocytic-monocytic lineage decision-making. Rather, our findings suggest that these transcription factors are only executing and reinforcing lineage choice once made. These results challenge the current prevailing model of early myeloid lineage choice.


Assuntos
Diferenciação Celular , Linhagem da Célula , Fator de Transcrição GATA1/metabolismo , Células Mieloides/citologia , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Eritrócitos/citologia , Retroalimentação Fisiológica , Feminino , Genes Reporter , Granulócitos/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Masculino , Megacariócitos/citologia , Camundongos , Modelos Biológicos , Monócitos/citologia , Reprodutibilidade dos Testes , Análise de Célula Única , Processos Estocásticos
11.
Nat Commun ; 7: 11903, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27301576

RESUMO

Stem cell identity depends on the integration of extrinsic and intrinsic signals, which directly influence the maintenance of their epigenetic state. Although Myc transcription factors play a major role in stem cell self-renewal and pluripotency, their integration with signalling pathways and epigenetic regulators remains poorly defined. We addressed this point by profiling the gene expression and epigenetic pattern in ESCs whose growth depends on conditional Myc activity. Here we show that Myc potentiates the Wnt/ß-catenin signalling pathway, which cooperates with the transcriptional regulatory network in sustaining ESC self-renewal. Myc activation results in the transcriptional repression of Wnt antagonists through the direct recruitment of PRC2 on these targets. The consequent potentiation of the autocrine Wnt/ß-catenin signalling induces the transcriptional activation of the endogenous Myc family members, which in turn activates a Myc-driven self-reinforcing circuit. Thus, our data unravel a Myc-dependent self-propagating epigenetic memory in the maintenance of ESC self-renewal capacity.


Assuntos
Redes Reguladoras de Genes/genética , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Autorrenovação Celular/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Retroalimentação Fisiológica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Fator Inibidor de Leucemia/farmacologia , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Proteínas do Grupo Polycomb/metabolismo , Transcrição Gênica/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
12.
Nat Cell Biol ; 17(10): 1235-46, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26389663

RESUMO

Transcription factor (TF) networks are thought to regulate embryonic stem cell (ESC) pluripotency. However, TF expression dynamics and regulatory mechanisms are poorly understood. We use reporter mouse ESC lines allowing non-invasive quantification of Nanog or Oct4 protein levels and continuous long-term single-cell tracking and quantification over many generations to reveal diverse TF protein expression dynamics. For cells with low Nanog expression, we identified two distinct colony types: one re-expressed Nanog in a mosaic pattern, and the other did not re-express Nanog over many generations. Although both expressed pluripotency markers, they exhibited differences in their TF protein correlation networks and differentiation propensities. Sister cell analysis revealed that differences in Nanog levels are not necessarily accompanied by differences in the expression of other pluripotency factors. Thus, regulatory interactions of pluripotency TFs are less stringently implemented in individual self-renewing ESCs than assumed at present.


Assuntos
Células-Tronco Embrionárias/metabolismo , Redes Reguladoras de Genes , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Diferenciação Celular/genética , Rastreamento de Células/métodos , Células Cultivadas , Células-Tronco Embrionárias/citologia , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia de Fluorescência , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Análise de Célula Única/métodos , Imagem com Lapso de Tempo/métodos , Fatores de Transcrição/metabolismo , Transdução Genética , Proteína Vermelha Fluorescente
13.
Integr Biol (Camb) ; 7(2): 178-83, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25514157

RESUMO

The kinetics of stem and progenitor cell differentiation at the single-cell level provides essential clues to the complexity of the underlying decision-making circuits. In many hematopoietic progenitor cells, differentiation is accompanied by the expression of lineage-specific markers and by a transition from a non-adherent to an adherent state. Here, using the granulocyte-macrophage progenitor (GMP) as a model, we introduce a label-free approach that allows one to follow the course of this transition in hundreds of single cells in parallel. We trap single cells in patterned arrays of micro-wells and use phase-contrast time-lapse movies to distinguish non-adherent from adherent cells by an analysis of Brownian motion. This approach allowed us to observe the kinetics of induced differentiation of primary bone-marrow-derived GMPs into macrophages. The time lapse started 2 hours after addition of the cytokine M-CSF, and nearly 80% of the population had accomplished the transition within the first 20 h. The analysis of Brownian motion proved to be a sensitive and robust tool for monitoring the transition, and thus provides a high-throughput method for the study of cell differentiation at the single-cell level.


Assuntos
Células Progenitoras de Granulócitos e Macrófagos/citologia , Dispositivos Lab-On-A-Chip , Animais , Fenômenos Biofísicos , Adesão Celular , Diferenciação Celular , Linhagem da Célula , Desenho de Equipamento , Células Progenitoras de Granulócitos e Macrófagos/fisiologia , Camundongos , Microscopia de Contraste de Fase , Movimento (Física) , Análise de Célula Única/instrumentação , Imagem com Lapso de Tempo/instrumentação
14.
Cell Stem Cell ; 15(5): 546-58, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25517464

RESUMO

Understanding the molecular control of cell fates is central to stem cell research. Such insight requires quantification of molecular and cellular behavior at the single-cell level. Recent advances now permit high-throughput molecular readouts from single cells as well as continuous, noninvasive observation of cell behavior over time. Here, we review current state-of-the-art approaches used to query stem cell fate at the single-cell level, including advances in lineage tracing, time-lapse imaging, and molecular profiling. We also offer our perspective on the advantages and drawbacks of available approaches, key technical limitations, considerations for data interpretation, and future innovation.


Assuntos
Análise de Célula Única/métodos , Pesquisa com Células-Tronco , Animais , Linhagem da Célula , Técnicas Genéticas , Humanos , Imagem Molecular , Transplante de Células-Tronco
15.
Exp Cell Res ; 329(2): 207-13, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25046868

RESUMO

Hematopoiesis is the cumulative consequence of finely tuned signaling pathways activated through extrinsic factors, such as local niche signals and systemic hematopoietic cytokines. Whether extrinsic factors actively instruct the lineage choice of hematopoietic stem and progenitor cells or are only selectively allowing survival and proliferation of already intrinsically lineage-committed cells has been debated over decades. Recent results demonstrated that cytokines can instruct lineage choice. However, the precise function of individual cytokine-triggered signaling molecules in inducing cellular events like proliferation, lineage choice, and differentiation remains largely elusive. Signal transduction pathways activated by different cytokine receptors are highly overlapping, but support the production of distinct hematopoietic lineages. Cellular context, signaling dynamics, and the crosstalk of different signaling pathways determine the cellular response of a given extrinsic signal. New tools to manipulate and continuously quantify signaling events at the single cell level are therefore required to thoroughly interrogate how dynamic signaling networks yield a specific cellular response.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Citocinas/farmacologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos
16.
Mol Biotechnol ; 56(8): 697-713, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24677035

RESUMO

Mammalian cells can be reprogrammed into induced pluripotent stem cells (iPSCs), a valuable tool for in vitro disease modeling and regenerative medicine. These applications demand for iPSCs devoid of reprogramming factor transgenes, but current procedures for the derivation of transgene-free iPSCs are inefficient and cumbersome. Here, we describe a new approach for the simple derivation of transgene-free iPSCs by the sequential use of two DNA recombinases, C31 Integrase and Cre, to control the genomic insertion and excision of a single, non-viral reprogramming vector. We show that such transgene-free iPSCs exhibit gene expression profiles and pluripotent developmental potential comparable to genuine, blastocyst-derived embryonic stem cells. As shown by a reporter iPSC line for the differentiation into midbrain dopaminergic neurons, the dual recombinase approach offers a simple and efficient way to derive transgene-free iPSCs for studying disease mechanisms and cell replacement therapies.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Biotecnologia , Diferenciação Celular , Células Cultivadas , Reprogramação Celular/genética , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Feminino , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Células-Tronco Pluripotentes Induzidas/transplante , Integrases/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Transcriptoma
17.
J Clin Invest ; 123(1): 469-78, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23257354

RESUMO

The scaffold protein p62 (sequestosome 1; SQSTM1) is an emerging key molecular link among the metabolic, immune, and proliferative processes of the cell. Here, we report that adipocyte-specific, but not CNS-, liver-, muscle-, or myeloid-specific p62-deficient mice are obese and exhibit a decreased metabolic rate caused by impaired nonshivering thermogenesis. Our results show that p62 regulates energy metabolism via control of mitochondrial function in brown adipose tissue (BAT). Accordingly, adipocyte-specific p62 deficiency led to impaired mitochondrial function, causing BAT to become unresponsive to ß-adrenergic stimuli. Ablation of p62 leads to decreased activation of p38 targets, affecting signaling molecules that control mitochondrial function, such as ATF2, CREB, PGC1α, DIO2, NRF1, CYTC, COX2, ATP5ß, and UCP1. p62 ablation in HIB1B and BAT primary cells demonstrated that p62 controls thermogenesis in a cell-autonomous manner, independently of brown adipocyte development or differentiation. Together, our data identify p62 as a novel regulator of mitochondrial function and brown fat thermogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteínas de Choque Térmico/metabolismo , Mitocôndrias/metabolismo , Termogênese/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Adipócitos Marrons/citologia , Tecido Adiposo Marrom/citologia , Animais , Células Cultivadas , Proteínas de Choque Térmico/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Especificidade de Órgãos/genética , Proteína Sequestossoma-1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Ann N Y Acad Sci ; 1266: 18-27, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22901252

RESUMO

Functional heterogeneity within stem and progenitor cells has been shown to influence cell fate decisions. Similarly, intracellular signaling activated by external stimuli is highly heterogeneous and its spatiotemporal activity is linked to future cell behavior. To quantify these heterogeneous states and link them to future cell fates, it is important to observe cell populations continuously with single cell resolution. Live cell imaging in combination with fluorescent biosensors for signaling activity serves as a powerful tool to study cellular and molecular heterogeneity and the long-term biological effects of signaling. Here, we describe these methodologies, their advantages over classical approaches, and we illustrate how they could be applied to improve our understanding of the importance of heterogeneous cellular and molecular responses to external signaling cues.


Assuntos
Imagem Molecular/métodos , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Técnicas Biossensoriais , Linhagem da Célula , Citocinas/metabolismo , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Modelos Biológicos , Transdução de Sinais
19.
Pharmacogenet Genomics ; 19(10): 790-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19741568

RESUMO

OBJECTIVE: As a key player in modulating both human physiological and behavioural functions including anxiety, perception and in particular appetite, serotonin (5-hydroxytryptamine, 5-HT) is likely to be involved in the aetiology of eating disorders. Studies showing serotonin receptor type 3 (5-HT3) receptors to mediate food intake depression (anorexic response) have triggered our interest in investigating the putative role of variants in the 5-HT3 receptor genes, HTR3A and HTR3B, in the susceptibility to anorexia nervosa (AN) and bulimia nervosa (BN). METHODS: Two hundred and sixty-five patients with AN and 91 patients with BN as well as 191 healthy controls served as a pilot study group for mutational analysis by direct sequencing. Variants showing a significant association were subsequently genotyped in an independent Spanish cohort of 78 patients with AN and 119 patients with BN as well as 331 healthy controls for replication purposes. RESULTS: In the pilot study, we found the coding HTR3B variant, p.Y129S, (rs1176744, P = 0.004, odds ratio = 2.06) to be associated with the restrictive subtype of AN. The association was confirmed in the Spanish study group (P = 0.034, odds ratio = 2.26). CONCLUSION: Our study provides first evidence for an involvement of 5-HT3 variants in the aetiopathology of eating disorders in humans.


Assuntos
Transtornos da Alimentação e da Ingestão de Alimentos/genética , Variação Genética , Receptores 5-HT3 de Serotonina/genética , Adolescente , Feminino , Humanos , Masculino , Modelos Biológicos , Projetos Piloto , Receptores de Serotonina/genética , Receptores 5-HT3 de Serotonina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...