Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell ; 156(4): 649-62, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24486105

RESUMO

Reprogramming somatic cells to induced pluripotency by Yamanaka factors is usually slow and inefficient and is thought to be a stochastic process. We identified a privileged somatic cell state, from which acquisition of pluripotency could occur in a nonstochastic manner. Subsets of murine hematopoietic progenitors are privileged whose progeny cells predominantly adopt the pluripotent fate with activation of endogenous Oct4 locus after four to five divisions in reprogramming conditions. Privileged cells display an ultrafast cell cycle of ∼8 hr. In fibroblasts, a subpopulation cycling at a similar ultrafast speed is observed after 6 days of factor expression and is increased by p53 knockdown. This ultrafast cycling population accounts for >99% of the bulk reprogramming activity in wild-type or p53 knockdown fibroblasts. Our data demonstrate that the stochastic nature of reprogramming can be overcome in a privileged somatic cell state and suggest that cell-cycle acceleration toward a critical threshold is an important bottleneck for reprogramming. PAPERCLIP:


Assuntos
Reprogramação Celular , Células Progenitoras de Granulócitos e Macrófagos/citologia , Células-Tronco Pluripotentes Induzidas , Animais , Células da Medula Óssea , Diferenciação Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Genes p53 , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Camundongos
2.
Stem Cells ; 41(10): 907-915, 2023 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-37386941

RESUMO

The role of serum response factor (Srf), a central mediator of actin dynamics and mechanical signaling, in cell identity regulation is debated to be either a stabilizer or a destabilizer. We investigated the role of Srf in cell fate stability using mouse pluripotent stem cells. Despite the fact that serum-containing cultures yield heterogeneous gene expression, deletion of Srf in mouse pluripotent stem cells leads to further exacerbated cell state heterogeneity. The exaggerated heterogeneity is detectible not only as increased lineage priming but also as the developmentally earlier 2C-like cell state. Thus, pluripotent cells explore more variety of cellular states in both directions of development surrounding naïve pluripotency, a behavior that is constrained by Srf. These results support that Srf functions as a cell state stabilizer, providing rationale for its functional modulation in cell fate intervention and engineering.


Assuntos
Células-Tronco Pluripotentes , Fator de Resposta Sérica , Camundongos , Animais , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Células-Tronco Pluripotentes/metabolismo , Diferenciação Celular/genética , Actinas/metabolismo , Expressão Gênica
3.
Stem Cells ; 39(1): 43-54, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33075202

RESUMO

There is wide variability in the propensity of somatic cells to reprogram into pluripotency in response to the Yamanaka factors. How to segregate these variabilities to enrich for cells of specific traits that reprogram efficiently remains challenging. Here we report that the variability in reprogramming propensity is associated with the activity of the MKL1/SRF transcription factor and concurs with small cell size as well as rapid cell cycle. Reprogramming progressive cells can be prospectively identified by their low activity of a widely used synthetic promoter, CAG. CAGlow cells arise and expand during cell cycle acceleration in the early reprogramming culture of both mouse and human fibroblasts. Our work illustrates a molecular scenario underlying the distinct reprogramming propensities and demonstrates a convenient practical approach for their enrichment.


Assuntos
Técnicas de Reprogramação Celular , Reprogramação Celular , Regiões Promotoras Genéticas , Fatores de Transcrição , Animais , Camundongos , Camundongos Transgênicos , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
5.
Blood ; 129(11): 1491-1502, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28053194

RESUMO

The hematopoietic stem cell-enriched miR-125 family microRNAs (miRNAs) are critical regulators of hematopoiesis. Overexpression of miR-125a or miR-125b is frequent in human acute myeloid leukemia (AML), and the overexpression of these miRNAs in mice leads to expansion of hematopoietic stem cells accompanied by perturbed hematopoiesis with mostly myeloproliferative phenotypes. However, whether and how miR-125 family miRNAs cooperate with known AML oncogenes in vivo, and how the resultant leukemia is dependent on miR-125 overexpression, are not well understood. We modeled the frequent co-occurrence of miR-125b overexpression and MLL translocations by examining functional cooperation between miR-125b and MLL-AF9 By generating a knock-in mouse model in which miR-125b overexpression is controlled by doxycycline induction, we demonstrated that miR-125b significantly enhances MLL-AF9-driven AML in vivo, and the resultant leukemia is partially dependent on continued overexpression of miR-125b Surprisingly, miR-125b promotes AML cell expansion and suppresses apoptosis involving a non-cell-intrinsic mechanism. MiR-125b expression enhances VEGFA expression and production from leukemia cells, in part by suppressing TET2 Recombinant VEGFA recapitulates the leukemia-promoting effects of miR-125b, whereas knockdown of VEGFA or inhibition of VEGF receptor 2 abolishes the effects of miR-125b In addition, significant correlation between miR-125b and VEGFA expression is observed in human AMLs. Our data reveal cooperative and dependent relationships between miR-125b and the MLL oncogene in AML leukemogenesis, and demonstrate a miR-125b-TET2-VEGFA pathway in mediating non-cell-intrinsic leukemia-promoting effects by an oncogenic miRNA.


Assuntos
Leucemia Mieloide Aguda/etiologia , MicroRNAs/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Apoptose , Proliferação de Células , Regulação Leucêmica da Expressão Gênica , Técnicas de Introdução de Genes , Hematopoese , Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda/metabolismo , Camundongos , Proteína de Leucina Linfoide-Mieloide/genética
6.
Ann Plast Surg ; 83(6): e92-e95, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31246672

RESUMO

Fibrosis is a consequence of aberrant wound healing processes that can be debilitating for patients and often are associated with highly morbid disease processes. Myofibroblasts play an important role in determining an appropriate physiologic response to tissue injury or an excessive response leading to fibrosis. Specifically, "supermature" focal adhesions, α-smooth muscle actin, and the myocardin-related transcription factor/serum response factor pathway likely play a significant role in the differentiation and survival of myofibroblasts in fibrotic lesions. Thus, targeting each of these and disrupting their functioning could lead to the development of therapeutic options for patients suffering from fibrosis and other sequelae of dysregulated wound healing. In this paper, we review the current literature concerning the roles of these three constituents of fibrotic signaling pathways, work already done in attempting to regulate these processes, and discuss the potential of these biomolecular constituents as therapeutic targets in future translational research.


Assuntos
Fibrose/genética , Regulação da Expressão Gênica , Transdução de Sinais , Cicatrização/genética , Diferenciação Celular/genética , Sobrevivência Celular/genética , Células Cultivadas , Feminino , Fibrose/patologia , Humanos , Masculino , Miofibroblastos/citologia , Pesquisa Translacional Biomédica , Cicatrização/fisiologia
7.
Nature ; 464(7290): 852-7, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20305640

RESUMO

Mesenchymal cells contribute to the 'stroma' of most normal and malignant tissues, with specific mesenchymal cells participating in the regulatory niches of stem cells. By examining how mesenchymal osteolineage cells modulate haematopoiesis, here we show that deletion of Dicer1 specifically in mouse osteoprogenitors, but not in mature osteoblasts, disrupts the integrity of haematopoiesis. Myelodysplasia resulted and acute myelogenous leukaemia emerged that had acquired several genetic abnormalities while having intact Dicer1. Examining gene expression altered in osteoprogenitors as a result of Dicer1 deletion showed reduced expression of Sbds, the gene mutated in Schwachman-Bodian-Diamond syndrome-a human bone marrow failure and leukaemia pre-disposition condition. Deletion of Sbds in mouse osteoprogenitors induced bone marrow dysfunction with myelodysplasia. Therefore, perturbation of specific mesenchymal subsets of stromal cells can disorder differentiation, proliferation and apoptosis of heterologous cells, and disrupt tissue homeostasis. Furthermore, primary stromal dysfunction can result in secondary neoplastic disease, supporting the concept of niche-induced oncogenesis.


Assuntos
Osso e Ossos/patologia , Leucemia Mieloide Aguda/patologia , Síndromes Mielodisplásicas/patologia , Células-Tronco/patologia , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Osso e Ossos/metabolismo , Diferenciação Celular , Linhagem da Célula , Feminino , Deleção de Genes , Hematopoese/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Mesoderma/citologia , Camundongos , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patologia , Fenótipo , Proteínas/genética , Proteínas/metabolismo , Ribonuclease III/deficiência , Ribonuclease III/genética , Ribonuclease III/metabolismo , Sarcoma Mieloide/genética , Sarcoma Mieloide/metabolismo , Sarcoma Mieloide/patologia , Nicho de Células-Tronco/metabolismo , Nicho de Células-Tronco/patologia , Células-Tronco/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia
8.
Proc Natl Acad Sci U S A ; 109(41): 16636-41, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012470

RESUMO

Deregulation of microRNA (miRNA) expression can lead to cancer initiation and progression. However, limited information exists on the function of miRNAs in cancer maintenance. We examined these issues in the case of myeloproliferative diseases and neoplasms (MPN), a collection of hematopoietic neoplasms regarded as preleukemic, thereby representing early neoplastic states. We report here that microRNA-125a (miR-125a)-induced MPN display a complex manner of oncogene dependence. Following a gain-of-function genomics screen, we overexpressed candidate miR-125a in vivo, which led to phenotypes consistent with an atypical MPN characterized by leukocytosis, monocytosis, splenomegaly, and progressive anemia. The diseased MPN state could be recapitulated in a doxycycline-inducible mouse model. Upon doxycycline withdrawal, the primary MPN phenotypes rapidly resolved after the discontinuation of miR-125a overexpression. However, reinduction of miR-125a led to complex phenotypes, with some animals rapidly developing lethal anemia with extensive damages in the spleen. Forced expression of miR-125a resulted in elevated cellular tyrosine phosphorylation and hypersensitivity toward hematopoietic cytokines. Furthermore, we demonstrate that miR-125a targets multiple protein phosphatases. Our data demonstrate that miR-125a-induced MPN is addicted to its sustained overexpression, and highlight the complex nature of oncogenic miRNA dependence in an early neoplastic state.


Assuntos
Neoplasias da Medula Óssea/genética , MicroRNAs/genética , Transtornos Mieloproliferativos/genética , Oncogenes/genética , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Neoplasias da Medula Óssea/metabolismo , Neoplasias da Medula Óssea/patologia , Transplante de Medula Óssea , Linhagem Celular , Ensaio de Unidades Formadoras de Colônias , Doxiciclina/farmacologia , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-3/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Stem Cells ; 31(5): 895-905, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23335078

RESUMO

Discovery of the cellular and molecular mechanisms of induced pluripotency has been hampered by its low efficiency and slow kinetics. Here, we report an experimental system with multicolor time-lapse microscopy that permits direct observation of pluripotency induction at single cell resolution, with temporal intervals as short as 5 minutes. Using granulocyte-monocyte progenitors as source cells, we visualized nascent pluripotent cells that emerge from a hematopoietic state. We engineered a suite of image processing and analysis software to annotate the behaviors of the reprogramming cells, which revealed the highly dynamic cell-cell interactions associated with early reprogramming. We observed frequent cell migration, which can lead to sister colonies, satellite colonies, and colonies of mixed genetic makeup. In addition, we discovered a previously unknown morphologically distinct two-cell intermediate of reprogramming, which occurs prior to other reprogramming landmarks. By directly visualizing the reprogramming process with E-cadherin inhibition, we demonstrate that E-cadherin is required for proper cellular interactions from an early stage of reprogramming, including the two-cell intermediate. The detailed cell-cell interactions revealed by this imaging platform shed light on previously unappreciated early reprogramming dynamics. This experimental system could serve as a powerful tool to dissect the complex mechanisms of early reprogramming by focusing on the relevant but rare cells with superb temporal and spatial resolution.


Assuntos
Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Reprogramação Celular/fisiologia , Animais , Caderinas/antagonistas & inibidores , Caderinas/metabolismo , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Imagem com Lapso de Tempo/métodos
10.
Development ; 137(10): 1586-94, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20430743

RESUMO

A key question in the stem cell field is how to balance the slow cycling of stem cells with active organ growth. Recent studies of the hair follicle stem cell niche have shown that this can be achieved by organizing the stem cell niche into two compartments: one that engages in immediate, rapid new growth and one that contributes later to long-term growth that fuels hair regeneration. Based on these and other recent findings, we propose that several other adult stem cell niches, including those in the blood, intestine and brain, have a similar bi-compartmental organization and that stem cells might work cooperatively with their progeny to sustain tissue regeneration.


Assuntos
Células-Tronco Adultas/citologia , Compartimento Celular/fisiologia , Nicho de Células-Tronco/citologia , Nicho de Células-Tronco/fisiologia , Células-Tronco Adultas/fisiologia , Animais , Linhagem da Célula/fisiologia , Imunofluorescência/métodos , Regeneração Tecidual Guiada/métodos , Folículo Piloso/citologia , Folículo Piloso/ultraestrutura , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/ultraestrutura , Modelos Biológicos
11.
Proc Natl Acad Sci U S A ; 107(32): 14229-34, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20616003

RESUMO

MicroRNAs influence hematopoietic differentiation, but little is known about their effects on the stem cell state. Here, we report that the microRNA processing enzyme Dicer is essential for stem cell persistence in vivo and a specific microRNA, miR-125a, controls the size of the stem cell population by regulating hematopoietic stem/progenitor cell (HSPC) apoptosis. Conditional deletion of Dicer revealed an absolute dependence for the multipotent HSPC population in a cell-autonomous manner, with increased HSPC apoptosis in mutant animals. An evolutionarily conserved microRNA cluster containing miR-99b, let-7e, and miR-125a was preferentially expressed in long-term hematopoietic stem cells. MicroRNA miR-125a alone was capable of increasing the number of hematopoietic stem cells in vivo by more than 8-fold. This result was accomplished through a differentiation stage-specific reduction of apoptosis in immature hematopoietic progenitors, possibly through targeting multiple proapoptotic genes. Bak1 was directly down-regulated by miR-125a and expression of a 3'UTR-less Bak1 blocked miR-125a-induced hematopoietic expansion in vivo. These data demonstrate cell-state-specific regulation by microRNA and identify a unique microRNA functioning to regulate the stem cell pool size.


Assuntos
RNA Helicases DEAD-box/fisiologia , Endorribonucleases/fisiologia , Células-Tronco Hematopoéticas/citologia , MicroRNAs/fisiologia , Animais , Apoptose/genética , Contagem de Células , Diferenciação Celular , Regulação para Baixo , Homeostase , Camundongos , Dados de Sequência Molecular , Ribonuclease III , Proteína Killer-Antagonista Homóloga a bcl-2/genética
12.
Elife ; 122023 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-36912771

RESUMO

Acute lymphoblastic and myeloblastic leukemias (ALL and AML) have been known to modify the bone marrow microenvironment and disrupt non-malignant hematopoiesis. However, the molecular mechanisms driving these alterations remain poorly defined. Using mouse models of ALL and AML, here we show that leukemic cells turn off lymphopoiesis and erythropoiesis shortly after colonizing the bone marrow. ALL and AML cells express lymphotoxin α1ß2 and activate lymphotoxin beta receptor (LTßR) signaling in mesenchymal stem cells (MSCs), which turns off IL7 production and prevents non-malignant lymphopoiesis. We show that the DNA damage response pathway and CXCR4 signaling promote lymphotoxin α1ß2 expression in leukemic cells. Genetic or pharmacological disruption of LTßR signaling in MSCs restores lymphopoiesis but not erythropoiesis, reduces leukemic cell growth, and significantly extends the survival of transplant recipients. Similarly, CXCR4 blocking also prevents leukemia-induced IL7 downregulation and inhibits leukemia growth. These studies demonstrate that acute leukemias exploit physiological mechanisms governing hematopoietic output as a strategy for gaining competitive advantage.


Assuntos
Leucemia Mieloide Aguda , Células-Tronco Mesenquimais , Animais , Camundongos , Leucemia Mieloide Aguda/patologia , Receptor beta de Linfotoxina/metabolismo , Interleucina-7/metabolismo , Linfopoese , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Células-Tronco Mesenquimais/metabolismo , Microambiente Tumoral
13.
Trends Cell Biol ; 32(8): 669-680, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35337714

RESUMO

The large arrays of cell types in a multicellular organism are defined by their stereotypic size and/or morphology, and, for cells in vivo, by their anatomic positions. Historically, this identity-structure-function correlation was conceptualized as arising from distinct gene expression programs that dictate how cells appear and behave. However, a growing number of studies suggest that a cell's mechanical state is also an important determinant of its identity, both in lineage-committed cells and in pluripotent stem cells. Defining the mechanism by which mechanical inputs influence complex cellular programs remains an area of ongoing investigation. Here, we discuss how the cytoskeleton actively participates in instructing the response of the nucleus and genome to integrate mechanical and biochemical inputs, with a primary focus on the role of the actomyosin-LINC (linker of nucleoskeleton and cytoskeleton) complex axis.


Assuntos
Núcleo Celular , Citoesqueleto , Actomiosina/metabolismo , Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Mecanotransdução Celular/fisiologia , Microtúbulos/metabolismo
14.
Nat Commun ; 12(1): 7334, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34921133

RESUMO

The erythroid terminal differentiation program couples sequential cell divisions with progressive reductions in cell size. The erythropoietin receptor (EpoR) is essential for erythroblast survival, but its other functions are not well characterized. Here we use Epor-/- mouse erythroblasts endowed with survival signaling to identify novel non-redundant EpoR functions. We find that, paradoxically, EpoR signaling increases red cell size while also increasing the number and speed of erythroblast cell cycles. EpoR-regulation of cell size is independent of established red cell size regulation by iron. High erythropoietin (Epo) increases red cell size in wild-type mice and in human volunteers. The increase in mean corpuscular volume (MCV) outlasts the duration of Epo treatment and is not the result of increased reticulocyte number. Our work shows that EpoR signaling alters the relationship between cycling and cell size. Further, diagnostic interpretations of increased MCV should now include high Epo levels and hypoxic stress.


Assuntos
Ciclo Celular , Tamanho Celular , Eritrócitos/citologia , Eritrócitos/metabolismo , Eritropoese , Receptores da Eritropoetina/metabolismo , Adulto , Animais , Antígenos CD/metabolismo , Antígenos CD4/metabolismo , Diferenciação Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Embrião de Mamíferos/metabolismo , Eritroblastos/citologia , Eritroblastos/efeitos dos fármacos , Eritroblastos/metabolismo , Eritropoetina/administração & dosagem , Eritropoetina/farmacologia , Feminino , Feto/metabolismo , Voluntários Saudáveis , Humanos , Ferro/metabolismo , Fígado/embriologia , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Proteínas Serina-Treonina Quinases/metabolismo , Receptores da Transferrina/metabolismo , Reticulócitos/citologia , Reticulócitos/efeitos dos fármacos , Reticulócitos/metabolismo , Transdução de Sinais , Proteína bcl-X/metabolismo
15.
FEBS Lett ; 2020 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-32441778

RESUMO

The cell division cycle is the generational period of cellular growth and propagation. Cell cycle progression needs to be highly regulated to preserve genomic fidelity while increasing cell number. In multicellular organisms, the cell cycle must also coordinate with cell fate specification during development and tissue homeostasis. Altered cell cycle dynamics play a central role also in a number of pathophysiological processes. Thus, extensive effort has been made to define the biochemical machineries that execute the cell cycle and their regulation, as well as implementing more sensitive and accurate cell cycle measurements. Here, we review the available techniques for cell cycle analysis, revisiting the assumptions behind conventional population-based measurements and discussing new tools to better address cell cycle heterogeneity in the single-cell era. We weigh the strengths, weaknesses, and trade-offs of methods designed to measure temporal aspects of the cell cycle. Finally, we discuss emerging techniques for capturing cell cycle speed at single-cell resolution in live animals.

16.
Stem Cell Reports ; 14(4): 730-743, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32243844

RESUMO

Yes-associated protein (YAP) is known to promote the stemness of multiple stem cell types, including pluripotent stem cells, while also antagonizing pluripotency during early embryogenesis. How YAP accomplishes these distinct functions remains unclear. Here, we report that, depending on the specific cells in which it is expressed, YAP could exhibit opposing effects on pluripotency induction from mouse somatic cells. Specifically, YAP inhibits pluripotency induction cell-autonomously but promotes it non-cell-autonomously. For its non-cell-autonomous role, YAP alters the expression of many secreted and matricellular proteins, including CYR61. YAP's non-cell-autonomous promoting effect could be recapitulated by recombinant CYR61 and abrogated by CYR61 depletion. Thus, we define a YAP-driven effect on enhancing pluripotency induction largely mediated by CYR61. Our work highlights the importance of considering the distinct contributions from heterologous cell types in deciphering cell fate control mechanisms and calls for careful re-examination of the co-existing bystander cells in complex cultures and tissues.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Células Cultivadas , Proteína Rica em Cisteína 61/metabolismo , Citocinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Ligação Proteica , Proteínas de Sinalização YAP
17.
Cell Rep ; 31(12): 107804, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32579930

RESUMO

Cell proliferation changes concomitantly with fate transitions during reprogramming, differentiation, regeneration, and oncogenesis. Methods to resolve cell cycle length heterogeneity in real time are currently lacking. Here, we describe a genetically encoded fluorescent reporter that captures live-cell cycle speed using a single measurement. This reporter is based on the color-changing fluorescent timer (FT) protein, which emits blue fluorescence when newly synthesized before maturing into a red fluorescent protein. We generated a mouse strain expressing an H2B-FT fusion reporter from a universally active locus and demonstrate that faster cycling cells can be distinguished from slower cycling ones on the basis of the intracellular fluorescence ratio between the FT's blue and red states. Using this reporter, we reveal the native cell cycle speed distributions of fresh hematopoietic cells and demonstrate its utility in analyzing cell proliferation in solid tissues. This system is broadly applicable for dissecting functional heterogeneity associated with cell cycle dynamics in complex tissues.


Assuntos
Ciclo Celular , Genes Reporter , Animais , Divisão Celular , Proliferação de Células , Células Cultivadas , Células-Tronco Hematopoéticas/metabolismo , Histonas/metabolismo , Proteínas Luminescentes , Camundongos , Modelos Biológicos , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteína Vermelha Fluorescente
18.
PLoS One ; 15(2): e0228651, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32045462

RESUMO

A new computational framework for FLow cytometric Analysis of Rare Events (FLARE) has been developed specifically for fast and automatic identification of rare cell populations in very large samples generated by platforms like multi-parametric flow cytometry. Using a hierarchical Bayesian model and information-sharing via parallel computation, FLARE rapidly explores the high-dimensional marker-space to detect highly rare populations that are consistent across multiple samples. Further it can focus within specified regions of interest in marker-space to detect subpopulations with desired precision.


Assuntos
Citometria de Fluxo/métodos , Modelos Teóricos , Automação Laboratorial/métodos , Probabilidade
19.
Nat Commun ; 11(1): 681, 2020 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-31996673

RESUMO

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

20.
Cancer Res ; 67(12): 5763-70, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575143

RESUMO

Previously, we showed that the bioactive green tea polyphenol epigallocatechin-3-gallate (EGCG) inhibits growth in soft agar of breast cancer cells with Her-2/neu overexpression. Using gene expression profiling, here we show that EGCG treatment of Her-2/neu-driven mammary tumor cells alters the expression of key regulators in the epithelial to mesenchymal transition (EMT) pathway, reducing invasive phenotype. Specifically, the epithelial genes E-cadherin, gamma-catenin, MTA3, and estrogen receptor alpha (ERalpha) were up-regulated by EGCG, whereas the proinvasive snail gene was down-regulated. Consistently, EGCG inhibited branching colony growth and invasion in Matrigel. EGCG treatment similarly inhibited invasive phenotype of mouse mammary tumor cells driven by Nuclear Factor-kappaB c-Rel and protein kinase CK2, frequently found overexpressed in human breast disease. Recently, we identified the Forkhead box O transcription factor FOXO3a as a major transcriptional regulator of ERalpha. Given the pivotal role of ERalpha in preventing EMT, we hypothesized that the activation of FOXO3a by EGCG plays an important role in the observed reversal of invasive phenotype in ERalpha-positive breast cancer cells. EGCG treatment activated FOXO3a. Ectopic expression of a constitutively active FOXO3a overrode transforming growth factor-beta1-mediated invasive phenotype and induced a more epithelial phenotype, which was dependent on ERalpha expression and signaling. Conversely, a dominant negative FOXO3a reduced epithelial phenotype of ERalpha-low breast cancer cells. These results identify, for the first time, a role for FOXO3a in the inhibition of invasive phenotype in breast cancer cells with active ERalpha signaling and elucidate a novel mechanism whereby EGCG represses EMT of breast cancer cells.


Assuntos
Anticarcinógenos/farmacologia , Neoplasias da Mama/patologia , Catequina/análogos & derivados , Receptor alfa de Estrogênio/efeitos dos fármacos , Fatores de Transcrição Forkhead/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Animais , Bebidas , Caderinas/efeitos dos fármacos , Caderinas/genética , Caderinas/metabolismo , Catequina/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Flavonoides/farmacologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Humanos , Immunoblotting , Invasividade Neoplásica/genética , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fenóis/farmacologia , Fenótipo , Polifenóis , Receptor ErbB-2/efeitos dos fármacos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , gama Catenina/efeitos dos fármacos , gama Catenina/genética , gama Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA