Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
N Engl J Med ; 389(9): 820-832, 2023 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-37646679

RESUMO

BACKGROUND: Sickle cell disease is caused by a defect in the ß-globin subunit of adult hemoglobin. Sickle hemoglobin polymerizes under hypoxic conditions, producing deformed red cells that hemolyze and cause vaso-occlusion that results in progressive organ damage and early death. Elevated fetal hemoglobin levels in red cells protect against complications of sickle cell disease. OTQ923, a clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-edited CD34+ hematopoietic stem- and progenitor-cell (HSPC) product, has a targeted disruption of the HBG1 and HBG2 (γ-globin) gene promoters that increases fetal hemoglobin expression in red-cell progeny. METHODS: We performed a tiling CRISPR-Cas9 screen of the HBG1 and HBG2 promoters by electroporating CD34+ cells obtained from healthy donors with Cas9 complexed with one of 72 guide RNAs, and we assessed the fraction of fetal hemoglobin-immunostaining erythroblasts (F cells) in erythroid-differentiated progeny. The gRNA resulting in the highest level of F cells (gRNA-68) was selected for clinical development. We enrolled participants with severe sickle cell disease in a multicenter, phase 1-2 clinical study to assess the safety and adverse-effect profile of OTQ923. RESULTS: In preclinical experiments, CD34+ HSPCs (obtained from healthy donors and persons with sickle cell disease) edited with CRISPR-Cas9 and gRNA-68 had sustained on-target editing with no off-target mutations and produced high levels of fetal hemoglobin after in vitro differentiation or xenotransplantation into immunodeficient mice. In the study, three participants received autologous OTQ923 after myeloablative conditioning and were followed for 6 to 18 months. At the end of the follow-up period, all the participants had engraftment and stable induction of fetal hemoglobin (fetal hemoglobin as a percentage of total hemoglobin, 19.0 to 26.8%), with fetal hemoglobin broadly distributed in red cells (F cells as a percentage of red cells, 69.7 to 87.8%). Manifestations of sickle cell disease decreased during the follow-up period. CONCLUSIONS: CRISPR-Cas9 disruption of the HBG1 and HBG2 gene promoters was an effective strategy for induction of fetal hemoglobin. Infusion of autologous OTQ923 into three participants with severe sickle cell disease resulted in sustained induction of red-cell fetal hemoglobin and clinical improvement in disease severity. (Funded by Novartis Pharmaceuticals; ClinicalTrials.gov number, NCT04443907.).


Assuntos
Anemia Falciforme , Sistemas CRISPR-Cas , Eritrócitos , Hemoglobina Fetal , Transplante de Células-Tronco Hematopoéticas , Animais , Camundongos , Anemia Falciforme/genética , Anemia Falciforme/terapia , Antígenos CD34 , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Hemoglobina Falciforme , Regiões Promotoras Genéticas
2.
Blood ; 136(11): 1303-1316, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32458004

RESUMO

Metabolic alterations in cancer represent convergent effects of oncogenic mutations. We hypothesized that a metabolism-restricted genetic screen, comparing normal primary mouse hematopoietic cells and their malignant counterparts in an ex vivo system mimicking the bone marrow microenvironment, would define distinctive vulnerabilities in acute myeloid leukemia (AML). Leukemic cells, but not their normal myeloid counterparts, depended on the aldehyde dehydrogenase 3a2 (Aldh3a2) enzyme that oxidizes long-chain aliphatic aldehydes to prevent cellular oxidative damage. Aldehydes are by-products of increased oxidative phosphorylation and nucleotide synthesis in cancer and are generated from lipid peroxides underlying the non-caspase-dependent form of cell death, ferroptosis. Leukemic cell dependence on Aldh3a2 was seen across multiple mouse and human myeloid leukemias. Aldh3a2 inhibition was synthetically lethal with glutathione peroxidase-4 (GPX4) inhibition; GPX4 inhibition is a known trigger of ferroptosis that by itself minimally affects AML cells. Inhibiting Aldh3a2 provides a therapeutic opportunity and a unique synthetic lethality to exploit the distinctive metabolic state of malignant cells.


Assuntos
Aldeído Oxirredutases/fisiologia , Carbolinas/farmacologia , Cicloexilaminas/farmacologia , Ferroptose/efeitos dos fármacos , Hematopoese/fisiologia , Leucemia Mieloide Aguda/enzimologia , Proteínas de Neoplasias/fisiologia , Fenilenodiaminas/farmacologia , Aldeído Oxirredutases/genética , Aldeídos/farmacologia , Animais , Linhagem Celular Tumoral , Citarabina/administração & dosagem , Doxorrubicina/administração & dosagem , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Peroxidação de Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína de Leucina Linfoide-Mieloide/fisiologia , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Ácido Oleico/farmacologia , Proteínas de Fusão Oncogênica/fisiologia , Oxirredução , Estresse Oxidativo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/antagonistas & inibidores , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/fisiologia
3.
Sci Rep ; 8(1): 16304, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30389991

RESUMO

CRISPR/Cas9 mediated gene editing of patient-derived hematopoietic stem and progenitor cells (HSPCs) ex vivo followed by autologous transplantation of the edited HSPCs back to the patient can provide a potential cure for monogenic blood disorders such as ß-hemoglobinopathies. One challenge for this strategy is efficient delivery of the ribonucleoprotein (RNP) complex, consisting of purified Cas9 protein and guide RNA, into HSPCs. Because ß-hemoglobinopathies are most prevalent in developing countries, it is desirable to have a reliable, efficient, easy-to-use and cost effective delivery method. With this goal in mind, we developed TRansmembrane Internalization Assisted by Membrane Filtration (TRIAMF), a new method to quickly and effectively deliver RNPs into HSPCs by passing a RNP and cell mixture through a filter membrane. We achieved robust gene editing in HSPCs using TRIAMF and demonstrated that the multilineage colony forming capacities and the competence for engraftment in immunocompromised mice of HSPCs were preserved post TRIAMF treatment. TRIAMF is a custom designed system using inexpensive components and has the capacity to process HSPCs at clinical scale.


Assuntos
Hemoglobina Fetal/genética , Filtração/métodos , Edição de Genes/métodos , Transplante de Células-Tronco Hematopoéticas , Ribonucleoproteínas/genética , Animais , Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas/genética , Células Cultivadas , Eletroporação/métodos , Feminino , Hemoglobina Fetal/metabolismo , Filtração/economia , Filtração/instrumentação , Terapia Genética/economia , Terapia Genética/instrumentação , Terapia Genética/métodos , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Humanos , Membranas Artificiais , Camundongos , Modelos Animais , RNA Guia de Cinetoplastídeos/genética , Transplante Autólogo
5.
Cell ; 167(5): 1310-1322.e17, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27863245

RESUMO

Stem cells determine homeostasis and repair of many tissues and are increasingly recognized as functionally heterogeneous. To define the extent of-and molecular basis for-heterogeneity, we overlaid functional, transcriptional, and epigenetic attributes of hematopoietic stem cells (HSCs) at a clonal level using endogenous fluorescent tagging. Endogenous HSC had clone-specific functional attributes over time in vivo. The intra-clonal behaviors were highly stereotypic, conserved under the stress of transplantation, inflammation, and genotoxic injury, and associated with distinctive transcriptional, DNA methylation, and chromatin accessibility patterns. Further, HSC function corresponded to epigenetic configuration but not always to transcriptional state. Therefore, hematopoiesis under homeostatic and stress conditions represents the integrated action of highly heterogeneous clones of HSC with epigenetically scripted behaviors. This high degree of epigenetically driven cell autonomy among HSCs implies that refinement of the concepts of stem cell plasticity and of the stem cell niche is warranted.


Assuntos
Epigenômica , Células-Tronco Hematopoéticas/citologia , Animais , Linhagem da Célula , Células Clonais/citologia , Fluorescência , Hematopoese , Inflamação/patologia , Camundongos , Transcrição Gênica
6.
Stem Cell Reports ; 7(2): 220-35, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27453006

RESUMO

Bone marrow niches for hematopoietic progenitor cells are not well defined despite their critical role in blood homeostasis. We previously found that cells expressing osteocalcin, a marker of mature osteolineage cells, regulate the production of thymic-seeding T lymphoid progenitors. Here, using a selective cell deletion strategy, we demonstrate that a subset of mesenchymal cells expressing osterix, a marker of bone precursors in the adult, serve to regulate the maturation of early B lymphoid precursors by promoting pro-B to pre-B cell transition through insulin-like growth factor 1 (IGF-1) production. Loss of Osx(+) cells or Osx-specific deletion of IGF-1 led to a failure of B cell maturation and the impaired adaptive immune response. These data highlight the notion that bone marrow is a composite of specialized niches formed by pairings of specific mesenchymal cells with parenchymal stem or lineage committed progenitor cells, thereby providing distinctive functional units to regulate hematopoiesis.


Assuntos
Linfócitos B/citologia , Diferenciação Celular , Mesoderma/citologia , Animais , Linfócitos B/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Fator de Transcrição Sp7/metabolismo
7.
Curr Opin Hematol ; 23(4): 331-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27177311

RESUMO

PURPOSE OF REVIEW: The bone marrow niche is increasingly recognized as heterogeneous with specific subtypes of mesenchymal niche cells governing the development or homeostasis of selective parenchymal hematopoietic subsets. The present review outlines recent efforts in dissecting these microniches regulated by unique cell pairings within the bone marrow and provides an overview of how the bone marrow orchestrates multiple facets of hematopoiesis. RECENT FINDINGS: Recent advancement in technologies has significantly improved our understanding of the cellular and molecular constituents that contribute to regulation of hematopoiesis and to maintenance of the hematopoietic stem cells (HSCs). Transgenic mouse models that enable endogenous cell deletion or lineage tracing, coupled with advanced intravital microscopy has identified several mesenchymal cell types, including the osteolineage cells, megakaryocytes, macrophages, perivascular cells, and Schwann cells, to be indispensible regulators of hematopoiesis. These niche cells, when perturbed, each caused very specific hematopoietic consequences including impairment in B-cell maturation, T lineage development, erythropoiesis, and impact different aspects of HSC behavior such as quiescence, mobilization, and response to acute stress signals. SUMMARY: The emerging concept is that the bone marrow environment is composed of multiple microniches, each consisting of unique pairing of distinct supportive stromal cells with distinct hematopoietic subtypes to regulate a particular branch of hematopoietic cell process. The bone marrow can be viewed as a carrier with subcompartments tailored to support different hematopoietic activities.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Comunicação Celular , Ciclo Celular , Diferenciação Celular , Movimento Celular , Eritropoese , Células-Tronco Hematopoéticas/citologia , Humanos , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/metabolismo , Linfopoese , Macrófagos/metabolismo , Megacariócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Osteogênese , Estresse Fisiológico , Sistema Nervoso Simpático/fisiologia
8.
Genom Data ; 5: 100-2, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26484235

RESUMO

T cells are known to develop in the thymus. However, molecular events that control the transition from hematopoietic progenitor cells in the bone marrow to T precursor cells seeded in the thymus remained poorly defined. Our recent report showed that osteocalcin (Ocn)-expressing bone cells in the bone marrow have major impact on T cell immunity by regulating T progenitor development in the bone marrow (Yu et al., 2015) [1]. Selective endogenous depletion of Ocn(+) cells by inducible diphtheria toxin receptor expression (OcnCre;iDTR) led to reduction of T-competent common lymphoid progenitors (Ly6D(-) CLPs) in the bone marrow and loss of T cells in the thymus. Expression of the Notch ligand DLL4 by Ocn(+) cells in the bone marrow ensures the production of Ly6D(-) CLPs, and expression of chemotactic molecules CCR7 and PSGL1 to enable subsequent thymic seeding. These data indicate that specific mesenchymal cells in bone marrow provide key molecular drivers enforcing thymus-seeding progenitor generation and thereby directly link skeletal biology to the production of T cell based adaptive immunity. Here we present the transcriptome profiles of Ly6D(-) CLPs derived from Ocn(+) cells deleted mice (OcnCre(+);iDTR) compared to those derived from control littermates (OcnCre(-);iDTR). These data are publically available from NCBI Gene Expression Omnibus (GEO) with the accession number GSE66102.

9.
Genom Data ; 5: 318-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26484277

RESUMO

The bone marrow niche is recognized as a central player in maintaining and regulating the behavior of hematopoietic stem and progenitor cells. Specific gain-of and loss-of function experiments perturbing a range of osteolineage cells or their secreted proteins had been shown to affect stem cell maintenance (Calvi et al, 2003 [1]; Stier et al., 2005 [2]; Zhang et al., 2003 [3]; Nilsson et al., 2005 [4]; Greenbaum et al., 2013 [5]) and engraftment (Adam et al., 2006, 2009 [6,7]). We used specific in vivo cell deletion approaches to dissect the niche cell-parenchymal cell dependency in a complex bone marrow microenvironment. Endogenous deletion of osteocalcin-expressing (Ocn(+)) cells led to a loss of T immune cells (Yu et al., 2015 [8]. Ocn(+) cells express the Notch ligand DLL4 to communicate with T-competent progenitors, and thereby ensuring T precursor production and expression of chemotactic molecules on their cell surface for subsequent thymic seeding. In contrast, depletion of osterix-expressing (Osx(+)) osteoprogenitors led to reduced B immune cells. These distinct hematopoietic phenotypes suggest specific pairing of mesenchymal niche cells and parenchymal hematopoietic cells in the bone marrow to create unique functional units to support hematopoiesis. Here, we present the global gene expression profiles of these osteolineage subtypes utilizing a triple fluorescent transgenic mouse model (OsxCre(+);Rosa-mCh(+);Ocn:Topaz(+)) that labels Osx(+) cells red, Ocn(+) cells green, and Osx(+) Ocn(+) cells yellow. This system allows isolation of distinct osteolineage subsets within the same animal by flow cytometry. Array data that have been described in our study [8] are also publically available from NCBI Gene Expression Omnibus (GEO) with the accession number GSE66042. Differences in gene expression may correlate with functional difference in supporting hematopoiesis.

10.
Stem Cells ; 33(7): 2280-93, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25851125

RESUMO

Notch is long recognized as a signaling molecule important for stem cell self-renewal and fate determination. Here, we reveal a novel adhesive role of Notch-ligand engagement in hematopoietic stem and progenitor cells (HSPCs). Using mice with conditional loss of O-fucosylglycans on Notch EGF-like repeats important for the binding of Notch ligands, we report that HSPCs with faulty ligand binding ability display enhanced cycling accompanied by increased egress from the marrow, a phenotype mainly attributed to their reduced adhesion to Notch ligand-expressing stromal cells and osteoblastic cells and their altered occupation in osteoblastic niches. Adhesion to Notch ligand-bearing osteoblastic or stromal cells inhibits wild type but not O-fucosylglycan-deficient HSPC cycling, independent of RBP-JK -mediated canonical Notch signaling. Furthermore, Notch-ligand neutralizing antibodies induce RBP-JK -independent HSPC egress and enhanced HSPC mobilization. We, therefore, conclude that Notch receptor-ligand engagement controls HSPC quiescence and retention in the marrow niche that is dependent on O-fucosylglycans on Notch.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Receptores Notch/metabolismo , Nicho de Células-Tronco/genética , Células Estromais/metabolismo , Animais , Humanos , Camundongos , Transdução de Sinais
11.
J Exp Med ; 212(5): 759-74, 2015 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-25918341

RESUMO

Production of the cells that ultimately populate the thymus to generate α/ß T cells has been controversial, and their molecular drivers remain undefined. Here, we report that specific deletion of bone-producing osteocalcin (Ocn)-expressing cells in vivo markedly reduces T-competent progenitors and thymus-homing receptor expression among bone marrow hematopoietic cells. Decreased intrathymic T cell precursors and decreased generation of mature T cells occurred despite normal thymic function. The Notch ligand DLL4 is abundantly expressed on bone marrow Ocn(+) cells, and selective depletion of DLL4 from these cells recapitulated the thymopoietic abnormality. These data indicate that specific mesenchymal cells in bone marrow provide key molecular drivers enforcing thymus-seeding progenitor generation and thereby directly link skeletal biology to the production of T cell-based adaptive immunity.


Assuntos
Células da Medula Óssea/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas de Membrana/imunologia , Células-Tronco Mesenquimais/imunologia , Linfócitos T/imunologia , Timo/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células da Medula Óssea/citologia , Proteínas de Ligação ao Cálcio , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Transgênicos , Osteocalcina/genética , Osteocalcina/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Timo/citologia
12.
J Exp Med ; 211(12): 2341-9, 2014 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-25332287

RESUMO

Paradoxical to its importance for generating a diverse T cell repertoire, thymic function progressively declines throughout life. This process has been at least partially attributed to the effects of sex steroids, and their removal promotes enhanced thymopoiesis and recovery from immune injury. We show that one mechanism by which sex steroids influence thymopoiesis is through direct inhibition in cortical thymic epithelial cells (cTECs) of Delta-like 4 (Dll4), a Notch ligand crucial for the commitment and differentiation of T cell progenitors in a dose-dependent manner. Consistent with this, sex steroid ablation (SSA) led to increased expression of Dll4 and its downstream targets. Importantly, SSA induced by luteinizing hormone-releasing hormone (LHRH) receptor antagonism bypassed the surge in sex steroids caused by LHRH agonists, the gold standard for clinical ablation of sex steroids, thereby facilitating increased Dll4 expression and more rapid promotion of thymopoiesis. Collectively, these findings not only reveal a novel mechanism underlying improved thymic regeneration upon SSA but also offer an improved clinical strategy for successfully boosting immune function.


Assuntos
Hormônios Esteroides Gonadais/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas de Membrana/imunologia , Receptores Notch/imunologia , Transdução de Sinais/imunologia , Timócitos/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Benzamidas , Proteínas de Ligação ao Cálcio , Linhagem Celular , Di-Hidrotestosterona/farmacologia , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Citometria de Fluxo , Hormônios Esteroides Gonadais/antagonistas & inibidores , Células HEK293 , Antagonistas de Hormônios/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfopoese/efeitos dos fármacos , Linfopoese/imunologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitrilas , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Receptores Androgênicos/imunologia , Receptores LHRH/agonistas , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/imunologia , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Testosterona/sangue , Testosterona/imunologia , Timócitos/citologia , Timo/citologia , Timo/imunologia
13.
Blood ; 124(19): 2937-47, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25202142

RESUMO

The glycosyltransferase gene, Ext1, is essential for heparan sulfate production. Induced deletion of Ext1 selectively in Mx1-expressing bone marrow (BM) stromal cells, a known population of skeletal stem/progenitor cells, in adult mice resulted in marked changes in hematopoietic stem and progenitor cell (HSPC) localization. HSPC egressed from BM to spleen after Ext1 deletion. This was associated with altered signaling in the stromal cells and with reduced vascular cell adhesion molecule 1 production by them. Further, pharmacologic inhibition of heparan sulfate mobilized qualitatively more potent and quantitatively more HSPC from the BM than granulocyte colony-stimulating factor alone, including in a setting of granulocyte colony-stimulating factor resistance. The reduced presence of endogenous HSPC after Ext1 deletion was associated with engraftment of transfused HSPC without any toxic conditioning of the host. Therefore, inhibiting heparan sulfate production may provide a means for avoiding the toxicities of radiation or chemotherapy in HSPC transplantation for nonmalignant conditions.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Heparitina Sulfato/biossíntese , N-Acetilglucosaminiltransferases/metabolismo , Células Estromais/metabolismo , Condicionamento Pré-Transplante , Animais , Anticoagulantes/farmacologia , Ligação Competitiva/imunologia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Proteínas de Fluorescência Verde/genética , Heparina/farmacologia , Heparitina Sulfato/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Acetilglucosaminiltransferases/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Células Estromais/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
14.
Cell ; 158(6): 1309-1323, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25215489

RESUMO

The balance between oxidative and nonoxidative glucose metabolism is essential for a number of pathophysiological processes. By deleting enzymes that affect aerobic glycolysis with different potencies, we examine how modulating glucose metabolism specifically affects hematopoietic and leukemic cell populations. We find that a deficiency in the M2 pyruvate kinase isoform (PKM2) reduces the levels of metabolic intermediates important for biosynthesis and impairs progenitor function without perturbing hematopoietic stem cells (HSCs), whereas lactate dehydrogenase A (LDHA) deletion significantly inhibits the function of both HSCs and progenitors during hematopoiesis. In contrast, leukemia initiation by transforming alleles putatively affecting either HSCs or progenitors is inhibited in the absence of either PKM2 or LDHA, indicating that the cell-state-specific responses to metabolic manipulation in hematopoiesis do not apply to the setting of leukemia. This finding suggests that fine-tuning the level of glycolysis may be explored therapeutically for treating leukemia while preserving HSC function.


Assuntos
Glicólise , Hematopoese , Leucemia/metabolismo , Animais , Deleção de Genes , Células-Tronco Hematopoéticas/metabolismo , Humanos , Isoenzimas/metabolismo , L-Lactato Desidrogenase/metabolismo , Lactato Desidrogenase 5 , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Piruvato Quinase/genética , Piruvato Quinase/metabolismo
15.
Gene ; 538(2): 328-33, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24440290

RESUMO

Factor inhibiting ATF4-mediated transcription (FIAT) interacts with Nascent polypeptide associated complex and coregulator alpha (αNAC). In cultured osteoblastic cells, this interaction contributes to maximal FIAT-mediated inhibition of Osteocalcin (Ocn) gene transcription. We set out to demonstrate the physiological relevance of this interaction by altering gene dosage in compound Fiat and Naca (encoding αNAC) heterozygous mice. Compound Naca(+/-); Fiat(+/-) heterozygous animals were viable, developed normally, and exhibited no significant difference in body weight compared with control littermate genotypes. Animals with a single Fiat allele had reduced Fiat mRNA expression without changes in the expression of related family members. Expression of the osteocyte differentiation marker Dmp1 was elevated in compound heterozygotes. Static histomorphometry parameters were assessed at 8weeks of age using microcomputed tomography (µCT). Trabecular measurements were not different between genotypes. Cortical thickness and area were not affected by gene dosage, but we measured a significant increase in cortical porosity in compound heterozygous mice, without changes in biomechanical parameters. The bone phenotype of compound Naca(+/-); Fiat(+/-) heterozygotes confirms that FIAT and αNAC are part of a common genetic pathway and support a role for the FIAT/αNAC interaction in normal bone physiology.


Assuntos
Proteínas Correpressoras/genética , Dosagem de Genes , Chaperonas Moleculares/genética , Proteínas Nucleares/genética , Animais , Fenômenos Biomecânicos , Desenvolvimento Ósseo/genética , Osso e Ossos/fisiologia , Proteínas Correpressoras/metabolismo , Epistasia Genética , Feminino , Expressão Gênica , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/metabolismo , Fenótipo
16.
Ann N Y Acad Sci ; 1068: 131-42, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16831913

RESUMO

The basic domain-leucine zipper protein, activating transcription factor 4 (ATF4), was recently shown to control key aspects of osteoblast biology. ATF4 regulates the timely onset of osteoblast differentiation, the synthesis of type I collagen, and the transcription of the osteocalcin and RANKL (receptor activator of NFkappa-B ligand) genes. Accordingly, the levels and activity of ATF4 are under tight control through mechanisms that include protein stability and phosphorylation. We have uncovered yet another mode of inhibition of ATF4 through its interaction with the leucine zipper protein FIAT (Factor Inhibiting ATF4-mediated Transcription, also described as gamma-taxilin). FIAT/gamma-taxilin localizes to the nucleus in osteoblasts and dimerizes with ATF4 to form inactive dimers, because it does not contain a DNA-binding basic domain moiety. The interaction of FIAT/gamma-taxilin with ATF4 thus inhibits ATF4-mediated transcription. Transgenic mice overexpressing FIAT/gamma-taxilin show osteopenia and reduced expression of the ATF4 target gene, osteocalcin. Interestingly, FIAT/gamma-taxilin also interacts with the transcriptional co-activator alphaNAC (Nascent polypeptide associated complex And Coactivator alpha), suggesting alternative, non-mutually exclusive mechanisms contributing to the inhibition of ATF4-dependent osteocalcin gene transcription by FIAT/gamma-taxilin.


Assuntos
Fator 4 Ativador da Transcrição/antagonistas & inibidores , Densidade Óssea/fisiologia , Proteínas de Transporte/fisiologia , Proteínas Nucleares/fisiologia , Proteínas de Transporte Vesicular/fisiologia , Animais , Reabsorção Óssea/genética , Divisão Celular , Proteínas Correpressoras , Humanos , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/fisiologia , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/fisiologia , Proteínas de Transporte Vesicular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA