Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 538, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38225226

RESUMO

Hematopoietic stem cells (HSCs) are capable of regenerating the blood system, but the instructive cues that direct HSCs to regenerate particular lineages lost to the injury remain elusive. Here, we show that iron is increasingly taken up by HSCs during anemia and induces erythroid gene expression and regeneration in a Tet2-dependent manner. Lineage tracing of HSCs reveals that HSCs respond to hemolytic anemia by increasing erythroid output. The number of HSCs in the spleen, but not bone marrow, increases upon anemia and these HSCs exhibit enhanced proliferation, erythroid differentiation, iron uptake, and TET2 protein expression. Increased iron in HSCs promotes DNA demethylation and expression of erythroid genes. Suppressing iron uptake or TET2 expression impairs erythroid genes expression and erythroid differentiation of HSCs; iron supplementation, however, augments these processes. These results establish that the physiological level of iron taken up by HSCs has an instructive role in promoting erythroid-biased differentiation of HSCs.


Assuntos
Anemia , Dioxigenases , Humanos , Baço , Células-Tronco Hematopoéticas/metabolismo , Diferenciação Celular , Ferro/metabolismo , Anemia/metabolismo , Células Eritroides , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo
3.
Cell Stem Cell ; 29(3): 386-399.e7, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35108519

RESUMO

Deregulation of transcription is a hallmark of acute myeloid leukemia (AML) that drives oncogenic expression programs and presents opportunities for therapeutic targeting. By integrating comprehensive pan-cancer enhancer landscapes with genetic dependency mapping, we find that AML-enriched enhancers encode for more selective tumor dependencies. We hypothesized that this approach could identify actionable dependencies downstream of oncogenic driver events and discovered a MYB-regulated AML-enriched enhancer regulating SEPHS2, a key component of the selenoprotein production pathway. Using a combination of patient samples and mouse models, we show that this enhancer upregulates SEPHS2, promoting selenoprotein production and antioxidant function required for AML survival. SEPHS2 and other selenoprotein pathway genes are required for AML growth in vitro. SEPHS2 knockout and selenium dietary restriction significantly delay leukemogenesis in vivo with little effect on normal hematopoiesis. These data validate the utility of enhancer mapping in target identification and suggest that selenoprotein production is an actionable target in AML.


Assuntos
Leucemia Mieloide Aguda , Selênio , Animais , Carcinogênese/genética , Elementos Facilitadores Genéticos/genética , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Oncogenes , Selênio/uso terapêutico
4.
Stem Cell Reports ; 16(8): 2014-2028, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34242617

RESUMO

Histone variants contribute to the complexity of the chromatin landscape and play an integral role in defining DNA domains and regulating gene expression. The histone H3 variant H3.3 is incorporated into genic elements independent of DNA replication by its chaperone HIRA. Here we demonstrate that Hira is required for the self-renewal of adult hematopoietic stem cells (HSCs) and to restrain erythroid differentiation. Deletion of Hira led to rapid depletion of HSCs while differentiated hematopoietic cells remained largely unaffected. Depletion of HSCs after Hira deletion was accompanied by increased expression of bivalent and erythroid genes, which was exacerbated upon cell division and paralleled increased erythroid differentiation. Assessing H3.3 occupancy identified a subset of polycomb-repressed chromatin in HSCs that depends on HIRA to maintain the inaccessible, H3.3-occupied state for gene repression. HIRA-dependent H3.3 incorporation thus defines distinct repressive chromatin that represses erythroid differentiation of HSCs.


Assuntos
Células-Tronco Adultas/metabolismo , Proteínas de Ciclo Celular/genética , Diferenciação Celular/genética , Células Eritroides/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Chaperonas de Histonas/genética , Fatores de Transcrição/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Proteínas de Ciclo Celular/metabolismo , Autorrenovação Celular/genética , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Hematopoese/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA-Seq/métodos , Fatores de Transcrição/metabolismo
5.
Sci Adv ; 7(30)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34290089

RESUMO

Metabolic dysregulation underlies malignant phenotypes attributed to cancer stem cells, such as unlimited proliferation and differentiation blockade. Here, we demonstrate that NAD+ metabolism enables acute myeloid leukemia (AML) to evade apoptosis, another hallmark of cancer stem cells. We integrated whole-genome CRISPR screening and pan-cancer genetic dependency mapping to identify NAMPT and NMNAT1 as AML dependencies governing NAD+ biosynthesis. While both NAMPT and NMNAT1 were required for AML, the presence of NAD+ precursors bypassed the dependence of AML on NAMPT but not NMNAT1, pointing to NMNAT1 as a gatekeeper of NAD+ biosynthesis. Deletion of NMNAT1 reduced nuclear NAD+, activated p53, and increased venetoclax sensitivity. Conversely, increased NAD+ biosynthesis promoted venetoclax resistance. Unlike leukemia stem cells (LSCs) in both murine and human AML xenograft models, NMNAT1 was dispensable for hematopoietic stem cells and hematopoiesis. Our findings identify NMNAT1 as a previously unidentified therapeutic target that maintains NAD+ for AML progression and chemoresistance.


Assuntos
Leucemia Mieloide Aguda , Nicotinamida-Nucleotídeo Adenililtransferase , Animais , Apoptose/genética , Homeostase , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Camundongos , NAD/metabolismo , Células-Tronco Neoplásicas/metabolismo , Nicotinamida-Nucleotídeo Adenililtransferase/genética , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo
6.
Blood ; 134(24): 2183-2194, 2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31697807

RESUMO

Altered metabolism fuels 2 hallmark properties of cancer cells: unlimited proliferation and differentiation blockade. Adenosine monophosphate-activated protein kinase (AMPK) is a master regulator of bioenergetics crucial for glucose metabolism in acute myeloid leukemia (AML), and its inhibition delays leukemogenesis, but whether the metabolic function of AMPK alters the AML epigenome remains unknown. Here, we demonstrate that AMPK maintains the epigenome of MLL-rearranged AML by linking acetyl-coenzyme A (CoA) homeostasis to Bromodomain and Extra-Terminal domain (BET) protein recruitment to chromatin. AMPK deletion reduced acetyl-CoA and histone acetylation, displacing BET proteins from chromatin in leukemia-initiating cells. In both mouse and patient-derived xenograft AML models, treating with AMPK and BET inhibitors synergistically suppressed AML. Our results provide a therapeutic rationale to target AMPK and BET for AML therapy.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Acetilcoenzima A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Leucemia Mieloide Aguda/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação Leucêmica da Expressão Gênica , Histonas/metabolismo , Homeostase , Humanos , Leucemia Mieloide Aguda/etiologia , Leucemia Mieloide Aguda/patologia , Camundongos , Gradação de Tumores , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Blood ; 134(7): 614-625, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31270104

RESUMO

Oncogenic mutations confer on cells the ability to propagate indefinitely, but whether oncogenes alter the cell fate of these cells is unknown. Here, we show that the transcriptional regulator PRDM16s causes oncogenic fate conversion by transforming cells fated to form platelets and erythrocytes into myeloid leukemia stem cells (LSCs). Prdm16s expression in megakaryocyte-erythroid progenitors (MEPs), which normally lack the potential to generate granulomonocytic cells, caused AML by converting MEPs into LSCs. Prdm16s blocked megakaryocytic/erythroid potential by interacting with super enhancers and activating myeloid master regulators, including PU.1. A CRISPR dropout screen confirmed that PU.1 is required for Prdm16s-induced leukemia. Ablating PU.1 attenuated leukemogenesis and reinstated the megakaryocytic/erythroid potential of leukemic MEPs in mouse models and human AML with PRDM16 rearrangement. Thus, oncogenic PRDM16 s expression gives MEPs an LSC fate by activating myeloid gene regulatory networks.


Assuntos
Transformação Celular Neoplásica/patologia , Proteínas de Ligação a DNA/genética , Leucemia Mieloide Aguda/patologia , Células Progenitoras de Megacariócitos e Eritrócitos/patologia , Fatores de Transcrição/genética , Animais , Transformação Celular Neoplásica/genética , Regulação Leucêmica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Leucemia Mieloide Aguda/genética , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Translocação Genética
8.
Stem Cell Reports ; 13(3): 545-558, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31257132

RESUMO

Bone marrow stromal cells (BMSCs) that express high levels of stem cell factor (SCF) and CXC chemokine ligand 12 (CXCL12) are one crucial component of the hematopoietic stem cell (HSC) niche. While the secreted factors produced by BMSCs to support HSCs have been well described, little is known regarding the transcriptional regulators controlling the cell fate of BMSCs and thus indirectly maintaining HSCs. BMI1 is a polycomb group protein that regulates HSCs both cell intrinsically and extrinsically, but it is unknown in which cell type and how BMI1 functions to maintain HSCs extrinsically. Here we show that Bmi1 maintains HSCs by preventing adipogenic differentiation of BMSCs. Bmi1 is highly expressed in BMSCs but becomes downregulated upon adipogenic differentiation and during aging. Deleting Bmi1 from BMSCs increased marrow adipocytes, induced HSC quiescence and depletion, and impaired hematopoiesis. We found that BMI1 repressed multiple developmental programs in BMSCs by safeguarding the repressive epigenetic marks histone H2A ubiquitylation and H3 lysine 27 trimethylation. We identified a novel adipogenic program governed by Pax3, which BMI1 repressed in BMSCs. Our results establish Bmi1 as a critical regulator of BMSC cell fate that suppresses marrow adipogenesis to create a supportive niche for HSCs.


Assuntos
Adipócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Adipócitos/citologia , Adipogenia , Animais , Diferenciação Celular , Senescência Celular , Feminino , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Histonas/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/metabolismo , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Nicho de Células-Tronco
9.
Blood Adv ; 2(11): 1220-1228, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29848758

RESUMO

Characterization of hematopoietic stem cells (HSCs) has advanced largely owing to transplantation assays, in which the developmental potential of HSCs is assessed generally in nonhomeostatic conditions. These studies established that adult HSCs extensively contribute to multilineage hematopoietic regeneration upon transplantation. On the contrary, recent studies performing lineage tracing of HSCs under homeostatic conditions have shown that adult HSCs may contribute far less to steady-state hematopoiesis than would be anticipated based on transplantation assays. Here, we used 2 independent HSC-lineage-tracing models to examine the contribution of adult HSCs to steady-state hematopoiesis. We show that adult HSCs contribute robustly to steady-state hematopoiesis, exhibiting faster efflux toward the myeloid lineages compared with lymphoid lineages. Platelets were robustly labeled by HSCs, reaching the same level of labeling as HSCs by 1 year of chase. Our results support the view that adult HSCs contribute to the continuous influx of blood cells during steady-state hematopoiesis.


Assuntos
Células-Tronco Adultas/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Adultas/citologia , Animais , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Transgênicos
10.
Exp Hematol ; 64: 33-44.e5, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29751067

RESUMO

Recent advances in next-generation sequencing have identified novel mutations and revealed complex genetic architectures in human hematological malignancies. Moving forward, new methods to quickly generate animal models that recapitulate the complex genetics of human hematological disorders are needed to transform the genetic information to new therapies. Here, we used a ribonucleoprotein-based CRISPR/Cas9 system to model human clonal hematopoiesis of indeterminate potential and acute myeloid leukemia (AML). We edited multiple genes recurrently mutated in hematological disorders, including those encoding epigenetic regulators, transcriptional regulators, and signaling components in murine hematopoietic stem/progenitor cells. Tracking the clonal dynamics by sequencing the indels induced by CRISPR/Cas9 revealed clonal expansion in some recipient mice that progressed to AML initiated by leukemia-initiating cells. Our results establish that the CRISPR/Cas9-mediated multiplex mutagenesis can be used to engineer a variety of murine models of hematological malignancies with complex genetic architectures seen in human disease.


Assuntos
Sistemas CRISPR-Cas , Células Clonais/patologia , Edição de Genes/métodos , Células-Tronco Hematopoéticas/patologia , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/patologia , Animais , Transplante de Medula Óssea , Proteína 9 Associada à CRISPR , Modelos Animais de Doenças , Progressão da Doença , Feminino , Genes Neoplásicos , Humanos , Mutação INDEL , Leucemia Mieloide Aguda/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Pré-Leucemia/genética , Pré-Leucemia/patologia , Ribonucleoproteínas/genética , Organismos Livres de Patógenos Específicos
11.
J Vis Exp ; (134)2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29708546

RESUMO

Advances in the hematopoietic stem cell (HSCs) field have been aided by methods to genetically engineer primary progenitor cells as well as animal models. Complete gene ablation in HSCs required the generation of knockout mice from which HSCs could be isolated, and gene ablation in primary human HSCs was not possible. Viral transduction could be used for knock-down approaches, but these suffered from variable efficacy. In general, genetic manipulation of human and mouse hematopoietic cells was hampered by low efficiencies and extensive time and cost commitments. Recently, CRISPR/Cas9 has dramatically expanded the ability to engineer the DNA of mammalian cells. However, the application of CRISPR/Cas9 to hematopoietic cells has been challenging, mainly due to their low transfection efficiencies, the toxicity of plasmid-based approaches and the slow turnaround time of virus-based protocols. A rapid method to perform CRISPR/Cas9-mediated gene editing in murine and human hematopoietic stem and progenitor cells with knockout efficiencies of up to 90% is provided in this article. This approach utilizes a ribonucleoprotein (RNP) delivery strategy with a streamlined three-day workflow. The use of Cas9-sgRNA RNP allows for a hit-and-run approach, introducing no exogenous DNA sequences in the genome of edited cells and reducing off-target effects. The RNP-based method is fast and straightforward: it does not require cloning of sgRNAs, virus preparation or specific sgRNA chemical modification. With this protocol, scientists should be able to successfully generate knockouts of a gene of interest in primary hematopoietic cells within a week, including downtimes for oligonucleotide synthesis. This approach will allow a much broader group of users to adapt this protocol for their needs.


Assuntos
Sistemas CRISPR-Cas/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco/metabolismo , Animais , Edição de Genes , Humanos , Camundongos
12.
Elife ; 72018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29451493

RESUMO

Activation of the unfolded protein response (UPR) sustains protein homeostasis (proteostasis) and plays a fundamental role in tissue maintenance and longevity of organisms. Long-range control of UPR activation has been demonstrated in invertebrates, but such mechanisms in mammals remain elusive. Here, we show that the female sex hormone estrogen regulates the UPR in hematopoietic stem cells (HSCs). Estrogen treatment increases the capacity of HSCs to regenerate the hematopoietic system upon transplantation and accelerates regeneration after irradiation. We found that estrogen signals through estrogen receptor α (ERα) expressed in hematopoietic cells to activate the protective Ire1α-Xbp1 branch of the UPR. Further, ERα-mediated activation of the Ire1α-Xbp1 pathway confers HSCs with resistance against proteotoxic stress and promotes regeneration. Our findings reveal a systemic mechanism through which HSC function is augmented for hematopoietic regeneration.


Assuntos
Endorribonucleases/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Resposta a Proteínas não Dobradas , Animais , Células Cultivadas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Transdução de Sinais
13.
Cell Rep ; 17(5): 1453-1461, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27783956

RESUMO

Our understanding of the mechanisms that regulate hematopoietic stem/progenitor cells (HSPCs) has been advanced by the ability to genetically manipulate mice; however, germline modification is time consuming and expensive. Here, we describe fast, efficient, and cost-effective methods to directly modify the genomes of mouse and human HSPCs using the CRISPR/Cas9 system. Using plasmid and virus-free delivery of guide RNAs alone into Cas9-expressing HSPCs or Cas9-guide RNA ribonucleoprotein (RNP) complexes into wild-type cells, we have achieved extremely efficient gene disruption in primary HSPCs from mouse (>60%) and human (∼75%). These techniques enabled rapid evaluation of the functional effects of gene loss of Eed, Suz12, and DNMT3A. We also achieved homology-directed repair in primary human HSPCs (>20%). These methods will significantly expand applications for CRISPR/Cas9 technologies for studying normal and malignant hematopoiesis.


Assuntos
Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Células-Tronco Hematopoéticas/metabolismo , Animais , Reparo do DNA , Deleção de Genes , Técnicas de Inativação de Genes , Humanos , Antígenos Comuns de Leucócito/metabolismo , Camundongos
14.
Cell Stem Cell ; 17(5): 585-96, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26440282

RESUMO

How cancer cells adapt to metabolically adverse conditions in patients and strive to proliferate is a fundamental question in cancer biology. Here we show that AMP-activated protein kinase (AMPK), a metabolic checkpoint kinase, confers metabolic stress resistance to leukemia-initiating cells (LICs) and promotes leukemogenesis. Upon dietary restriction, MLL-AF9-induced murine acute myeloid leukemia (AML) activated AMPK and maintained leukemogenic potential. AMPK deletion significantly delayed leukemogenesis and depleted LICs by reducing the expression of glucose transporter 1 (Glut1), compromising glucose flux, and increasing oxidative stress and DNA damage. LICs were particularly dependent on AMPK to suppress oxidative stress in the hypoglycemic bone marrow environment. Strikingly, AMPK inhibition synergized with physiological metabolic stress caused by dietary restriction and profoundly suppressed leukemogenesis. Our results indicate that AMPK protects LICs from metabolic stress and that combining AMPK inhibition with physiological metabolic stress potently suppresses AML by inducing oxidative stress and DNA damage.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Medula Óssea/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células-Tronco Neoplásicas/metabolismo , Estresse Oxidativo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/deficiência , Animais , Medula Óssea/enzimologia , Medula Óssea/patologia , Dano ao DNA , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Baço/metabolismo , Baço/patologia
15.
Blood ; 123(21): 3344-53, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24713928

RESUMO

Thrombotic thrombocytopenic purpura (TTP) is a life-threatening disease that presents with thrombocytopenia, disseminated thrombosis, hemolytic anemia, and organ dysfunction. The etiology of TTP has revealed that patients share a deficiency in plasma protease a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), the enzyme responsible for cleaving ultra-large von Willebrand factor (VWF) multimers into nonthrombogenic fragments. Therefore, existing TTP mouse models were developed by targeted disruption of the ADAMTS13 gene. ADAMTS13(-/-) mice are mostly asymptomatic in the absence of a trigger, as redundant proteases appear to take on VWF processing. As an alternative approach to creating one such model, we devised a strategy based on the expression of a cleavage-resistant VWF mutant in mice. The creation of a disulfide bond within the A2 domain of VWF was found to render VWF multimers resistant to proteolysis by plasma proteases under flow. Furthermore, mice expressing the murine VWF/p.S1494C-p.A1534C mutant present with symptoms characteristics of acute TTP such as thrombocytopenia, red cell shredding, accumulation of VWF-rich thrombi in the microvasculature, and advanced TTP symptoms such as renal dysfunction and splenomegaly. Because this model appears to faithfully emulate the pathophysiology of TTP, it should prove most useful in the study of microangiopathic diseases and their treatment.


Assuntos
Púrpura Trombocitopênica Trombótica/genética , Púrpura Trombocitopênica Trombótica/metabolismo , Fator de von Willebrand/química , Fator de von Willebrand/genética , Sequência de Aminoácidos , Animais , Dissulfetos/química , Dissulfetos/metabolismo , Células HEK293 , Humanos , Hidrólise , Rim/metabolismo , Rim/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Peptídeo Hidrolases/metabolismo , Estrutura Terciária de Proteína , Púrpura Trombocitopênica Trombótica/patologia , Fator de von Willebrand/metabolismo
16.
Nature ; 505(7484): 555-8, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24451543

RESUMO

Sexually dimorphic mammalian tissues, including sexual organs and the brain, contain stem cells that are directly or indirectly regulated by sex hormones. An important question is whether stem cells also exhibit sex differences in physiological function and hormonal regulation in tissues that do not show sex-specific morphological differences. The terminal differentiation and function of some haematopoietic cells are regulated by sex hormones, but haematopoietic stem-cell function is thought to be similar in both sexes. Here we show that mouse haematopoietic stem cells exhibit sex differences in cell-cycle regulation by oestrogen. Haematopoietic stem cells in female mice divide significantly more frequently than in male mice. This difference depends on the ovaries but not the testes. Administration of oestradiol, a hormone produced mainly in the ovaries, increased haematopoietic stem-cell division in males and females. Oestrogen levels increased during pregnancy, increasing haematopoietic stem-cell division, haematopoietic stem-cell frequency, cellularity, and erythropoiesis in the spleen. Haematopoietic stem cells expressed high levels of oestrogen receptor-α (ERα). Conditional deletion of ERα from haematopoietic stem cells reduced haematopoietic stem-cell division in female, but not male, mice and attenuated the increases in haematopoietic stem-cell division, haematopoietic stem-cell frequency, and erythropoiesis during pregnancy. Oestrogen/ERα signalling promotes haematopoietic stem-cell self-renewal, expanding splenic haematopoietic stem cells and erythropoiesis during pregnancy.


Assuntos
Estrogênios/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Contagem de Células , Divisão Celular/efeitos dos fármacos , Eritropoese , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Gravidez , Caracteres Sexuais , Transdução de Sinais/efeitos dos fármacos , Baço/citologia
17.
Mol Cell Neurosci ; 46(1): 200-12, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20851768

RESUMO

The neurobiological mechanisms of emotional modulation and the molecular pathophysiology of anxiety disorders are largely unknown. The fibroblast growth factor (FGF) family has been implicated in the regulation of many physiological and pathological processes, which include the control of emotional behaviors. The present study examined mice with a targeted deletion of the fgf-bp3 gene, which encodes a novel FGF-binding protein, in animal models relevant to anxiety. To define the behavioral consequences of FGF-BP3 deficiency, we evaluated fgf-bp3-deficient mice using anxiety-related behavioral paradigms that provide a conflict between the desire to explore an unknown area or objects and the aversion to a brightly lit open space. The fgf-bp3-deficient mice exhibited alterations in time spent in the central area of the open-field arena, were less active in the lit areas of a light/dark transition test, and had a prolonged latency to feed during a novelty-induced hypophagia test. These changes were associated with alterations in light-induced orbitofrontal cortex (OFC) activation in an extracellular signal-regulated kinase (ERK) pathway-dependent manner. These results demonstrate that FGF-BP3 is a potent mediator of anxiety-related behaviors in mice and suggest that distinct pathways regulate emotional behaviors. Therefore, FGF-BP3 plays a critical role in the regulation of emotional states and in the development of anxiety disorders and should be investigated as a therapeutic target for anxiety disease in humans.


Assuntos
Ansiedade/metabolismo , Comportamento Animal/fisiologia , Proteínas de Transporte/metabolismo , Animais , Ansiedade/genética , Proteínas de Transporte/genética , Linhagem Celular , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Testes Neuropsicológicos
18.
Dev Dyn ; 239(8): 2233-45, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20568244

RESUMO

Using comprehensive genetic studies on neuronal stem/progenitors cells through genome-wide screening with oligonucleotide arrays, we identified an endoplasmic reticulum (ER) -resident protein, Tweety homologue 1 (ttyh1). Ttyh1 encodes a glycosylated protein composed of five predicted transmembrane segments and a C-terminus that is enriched in negatively charged residues capable of Ca(2+) binding. Ttyh1-containing membranes changed to segmented tubuloreticular structures during mitosis, suggesting that the ER-containing Ttyh1 could be responsible for Ca(2+) sequestration and Ca(2+) concentration regulation during mitosis. Ttyh1 inactivation in mice resulted in early embryonic lethality before organization of the nervous system, revealing that ttyh1 is essential in murine embryonic development. Our findings indicate that Ttyh1 plays an indispensable role during mitosis in early embryogenesis, possibly by maintaining Ca(2+) homeostasis in the ER.


Assuntos
Desenvolvimento Embrionário , Retículo Endoplasmático/química , Proteínas de Membrana/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Retículo Endoplasmático/metabolismo , Estudo de Associação Genômica Ampla , Proteínas de Membrana/análise , Camundongos , Mitose , Neurônios/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco/citologia
19.
Anim Sci J ; 81(2): 215-22, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20438503

RESUMO

The effect of L-carnitine supplementation on mitogen (concanavalin A, Con A) induced proliferation of mononuclear cells (MNC) in the spleen was investigated in broiler chickens at different ages. Day-old chickens were fed a diet supplemented with or without L-carnitine (100 ppm) for 24 days. The carnitine-supplemented group showed greater proliferation of MNC in the spleen in response to Con A than that of the control group at 24 days of age. In addition, at 24 days of age the carnitine-supplemented group showed higher expression of interleukin (IL)-2 and interferon (IFN)-gamma mRNA, but lower expression of inducible nitric oxide synthase (iNOS) in the Con A-stimulated splenic MNC than the control group. The enhancement effect of L-carnitine on MNC proliferation and IL-2 mRNA expression was not found in chicks at 14 days of age. Addition of L-carnitine (50 nmol/mL) to the culture medium enhanced proliferation and IL-2 mRNA expression of splenic MNC obtained from 24-day-old but not from 14-day-old broiler chickens. The results suggest that L-carnitine is capable of enhancing MNC proliferation in broiler chickens at 24 days of age partly through increasing IL-2 and IFN-gamma production and decreasing NO production.


Assuntos
Carnitina/administração & dosagem , Galinhas/imunologia , Leucócitos Mononucleares/metabolismo , Baço/citologia , Baço/imunologia , Envelhecimento , Ração Animal , Animais , Proliferação de Células/efeitos dos fármacos , Galinhas/metabolismo , Concanavalina A/imunologia , Imunoglobulina G/sangue , Interferon gama/imunologia , Interleucina-2/imunologia , Masculino , RNA Mensageiro/metabolismo , Linfócitos T/imunologia
20.
Mech Dev ; 125(5-6): 441-50, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18337066

RESUMO

Borealin/DasraB is a member of the chromosomal passenger protein complex (CPC) required for proper segregation of chromosomes during mitosis. In Drosophila melanogaster, inactivation of Borealin/DasraB results in polyploidy, delayed mitosis and abnormal tissue development, indicating its critical role for cell proliferation. However, the in vivo role of mammalian Borealin/DasraB remains unclear. Here, we analyzed the expression of Borealin/DasraB and found that borealin is widely expressed in embryonic tissues and later restricted to adult tissues which relies on rapid cell proliferation. To determine the role of borealin during mouse development, we generated borealin-null mice through targeted disruption. While heterozygous mice developed normally, disruption of both borealin alleles resulted in early embryonic lethality by 5.5 dpc (days postcoitus) due to mitotic defects and apoptosis in blastocyst cells that showed microtubule disorganization and no CPC enrichment. At 5.5 dpc, borealin-null embryos exhibited excessive apoptosis and elevated expression of p53. However, loss of p53 did not abrogate or delay embryonic lethality, revealing that Borealin/DasraB inactivation triggered impaired mitosis and apoptosis though p53-independent mechanisms. Our data show that Borealin/DasraB is essential for cell proliferation during early embryonic development, and its early embryonic lethality cannot be rescued by the loss of p53.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Blastocisto/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona , Biologia do Desenvolvimento/métodos , Genes p53 , Heterozigoto , Camundongos , Camundongos Transgênicos , Microtúbulos/metabolismo , Mitose , Modelos Genéticos , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Ploidias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...