Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Immunol ; 199(8): 2701-2712, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28931604

RESUMO

Total body irradiation (TBI) damages hematopoietic cells in the bone marrow and thymus; however, the long-term effects of irradiation with aging remain unclear. In this study, we found that the impact of radiation on thymopoiesis in mice varied by sex and dose but, overall, thymopoiesis remained suppressed for ≥12 mo after a single exposure. Male and female mice showed a long-term dose-dependent reduction in thymic cKit+ lymphoid progenitors that was maintained throughout life. Damage to hematopoietic stem cells (HSCs) in the bone marrow was dose dependent, with as little as 0.5 Gy causing a significant long-term reduction. In addition, the potential for T lineage commitment was radiation sensitive with aging. Overall, the impact of irradiation on the hematopoietic lineage was more severe in females. In contrast, the rate of decline in thymic epithelial cell numbers with age was radiation-sensitive only in males, and other characteristics including Ccl25 transcription were unaffected. Taken together, these data suggest that long-term suppression of thymopoiesis after sublethal irradiation was primarily due to fewer progenitors in the BM combined with reduced potential for T lineage commitment. A single irradiation dose also caused synchronization of thymopoiesis, with a periodic thymocyte differentiation profile persisting for at least 12 mo postirradiation. This study suggests that the number and capability of HSCs for T cell production can be dramatically and permanently damaged after a single relatively low TBI dose, accelerating aging-associated thymic involution. Our findings may impact evaluation and therapeutic intervention of human TBI events.


Assuntos
Células da Medula Óssea/fisiologia , Hematopoese/efeitos da radiação , Síndromes de Imunodeficiência/imunologia , Células Progenitoras Linfoides/fisiologia , Linfócitos T/fisiologia , Timo/efeitos da radiação , Envelhecimento , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Feminino , Síndromes de Imunodeficiência/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-kit/metabolismo , Timo/imunologia , Irradiação Corporal Total/efeitos adversos
2.
Proc Natl Acad Sci U S A ; 111(8): 2885-90, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24516138

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a medicinally important glycoprotein, used as an immunostimulant following bone-marrow transplant. On the basis of reports of its potential utility as an anticancer vaccine adjuvant, we undertook to develop a synthetic route toward single-glycoform GM-CSF. We describe herein a convergent total synthesis of GM-CSF aglycone and two homogeneous glycoforms. Analytical and biological studies confirm the structure and activity of these synthetic congeners.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/síntese química , Modelos Moleculares , Conformação Proteica , Alanina/química , Sequência de Aminoácidos , Cisteína/química , Escherichia coli , Glicosilação , Dados de Sequência Molecular , Estrutura Molecular
3.
J Am Chem Soc ; 137(40): 13167-75, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26401918

RESUMO

Human granulocyte colony-stimulating factor (G-CSF) is an endogenous glycoprotein involved in hematopoiesis. Natively glycosylated and nonglycosylated recombinant forms, lenograstim and filgrastim, respectively, are used clinically to manage neutropenia in patients undergoing chemotherapeutic treatment. Despite their comparable therapeutic potential, the purpose of O-linked glycosylation at Thr133 remains a subject of controversy. In light of this, we have developed a synthetic platform to prepare G-CSF aglycone with the goal of enabling access to native and designed glycoforms with site-selectivity and glycan homogeneity. To address the synthesis of a relatively large, aggregation-prone sequence, we advanced an isonitrile-mediated ligation method. The chemoselective activation and coupling of C-terminal peptidyl Gly thioacids with the N-terminus of an unprotected peptide provide ligated peptides directly in a manner complementary to that with conventional native chemical ligation-desulfurization strategies. Herein, we describe the details and application of this method as it enabled the convergent total synthesis of G-CSF aglycone.


Assuntos
Fator Estimulador de Colônias de Granulócitos/química , Peptídeos/química , Sequência de Aminoácidos , Eletroforese em Gel de Poliacrilamida , Dados de Sequência Molecular
4.
Blood ; 121(20): 4082-9, 2013 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-23520338

RESUMO

Direct transduction of the homeobox (HOX) protein HOXB4 promotes the proliferation of hematopoietic stem cells (HSCs) without induction of leukemogenesis, but requires frequent administration to overcome its short protein half-life (∼1 hour). We demonstrate here that HOXB4 protein levels are post-translationally regulated by the CUL4 ubiquitin ligase, and define the degradation signal sequence (degron) of HOXB4 required for CUL4-mediated destruction. Additional HOX paralogs share the conserved degron in the homeodomain and are also subject to CUL4-mediated degradation, indicating that CUL4 likely controls the stability of all HOX proteins. Moreover, we engineered a degradation-resistant HOXB4 that conferred a growth advantage over wild-type HOXB4 in myeloid progenitor cells. Direct transduction of recombinant degradation-resistant HOXB4 protein to human adult HSCs significantly enhanced their maintenance in a more primitive state both in vitro and in transplanted NOD/SCID/IL2R-γ(null) mice compared with transduction with wild-type HOXB4 protein. Our studies demonstrate the feasibility of engineering a stable HOXB4 variant to overcome a major technical hurdle in the ex vivo expansion of adult HSCs and early progenitors for human therapeutic use.


Assuntos
Células-Tronco Adultas/fisiologia , Proliferação de Células , Proteínas Culina/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Células-Tronco Adultas/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Proteínas Culina/genética , Proteínas Culina/metabolismo , Estudos de Viabilidade , Células HeLa , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Cultura Primária de Células/métodos , Engenharia de Proteínas , Proteólise , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
5.
Top Curr Chem ; 362: 1-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25805144

RESUMO

Glycoproteins are an important class of naturally occurring biomolecules which play a pivotal role in many biological processes. They are biosynthesized as complex mixtures of glycoforms through post-translational protein glycosylation. This fact, together with the challenges associated with producing them in homogeneous form, has hampered detailed structure-function studies of glycoproteins as well as their full exploitation as potential therapeutic agents. By contrast, chemical synthesis offers the unique opportunity to gain access to homogeneous glycoprotein samples for rigorous biological evaluation. Herein, we review recent methods for the assembly of complex glycopeptides and glycoproteins and present several examples from our laboratory towards the total chemical synthesis of clinically relevant glycosylated proteins that have enabled synthetic access to full-length homogeneous glycoproteins.


Assuntos
Glicoproteínas/síntese química , Glicosilação , Estrutura Molecular , Engenharia de Proteínas
6.
Nat Chem Biol ; 9(12): 840-848, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24161946

RESUMO

Efforts to develop more effective therapies for acute leukemia may benefit from high-throughput screening systems that reflect the complex physiology of the disease, including leukemia stem cells (LSCs) and supportive interactions with the bone marrow microenvironment. The therapeutic targeting of LSCs is challenging because LSCs are highly similar to normal hematopoietic stem and progenitor cells (HSPCs) and are protected by stromal cells in vivo. We screened 14,718 compounds in a leukemia-stroma co-culture system for inhibition of cobblestone formation, a cellular behavior associated with stem-cell function. Among those compounds that inhibited malignant cells but spared HSPCs was the cholesterol-lowering drug lovastatin. Lovastatin showed anti-LSC activity in vitro and in an in vivo bone marrow transplantation model. Mechanistic studies demonstrated that the effect was on target, via inhibition of HMG-CoA reductase. These results illustrate the power of merging physiologically relevant models with high-throughput screening.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Leucemia , Células-Tronco Neoplásicas/efeitos dos fármacos , Linhagem Celular Tumoral , Células-Tronco Hematopoéticas , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/fisiologia
7.
Proc Natl Acad Sci U S A ; 109(34): E2276-83, 2012 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-22652566

RESUMO

Most gastrointestinal stromal tumors (GISTs) harbor a gain-of-function mutation in the Kit receptor. GIST patients treated with the tyrosine kinase inhibitor imatinib frequently develop imatinib resistance as a result of second-site Kit mutations. To investigate the consequences of second-site Kit mutations on GIST development and imatinib sensitivity, we engineered a mouse model carrying in the endogenous Kit locus both the Kit(V558Δ) mutation found in a familial case of GIST and the Kit(T669I) (human KIT(T670I)) "gatekeeper" mutation found in imatinib-resistant GIST patients. Similar to Kit(V558/+) mice, Kit(V558;T669I/+) mice developed gastric and colonic interstitial cell of Cajal hyperplasia as well as cecal GIST. In contrast to the single-mutant Kit(V558/+) control mice, treatment of the Kit(V558;T669I/+) mice with either imatinib or dasatinib failed to inhibit oncogenic Kit signaling and GIST growth. However, this resistance could be overcome by treatment of Kit(V558;T669I/+) mice with sunitinib or sorafenib. Although tumor lesions were smaller in Kit(V558;T669I/+) mice than in single-mutant mice, both interstitial cell of Cajal hyperplasia and mast cell hyperplasia were exacerbated in Kit(V558;T669I/+) mice. Strikingly, the Kit(V558;T669I/+) mice developed a pronounced polycythemia vera-like erythrocytosis in conjunction with microcytosis. This mouse model should be useful for preclinical studies of drug candidates designed to overcome imatinib resistance in GIST and to investigate the consequences of oncogenic KIT signaling in hematopoietic as well as other cell lineages.


Assuntos
Eritrócitos/citologia , Tumores do Estroma Gastrointestinal/genética , Mutação , Piperazinas/farmacologia , Policitemia/genética , Proteínas Proto-Oncogênicas c-kit/genética , Pirimidinas/farmacologia , Animais , Antineoplásicos/farmacologia , Benzamidas , Linhagem da Célula , Dasatinibe , Modelos Animais de Doenças , Resistência a Medicamentos , Resistencia a Medicamentos Antineoplásicos/genética , Éxons , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Mesilato de Imatinib , Camundongos , Fenótipo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Tiazóis/farmacologia
8.
bioRxiv ; 2023 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-37034724

RESUMO

Transition between activation and quiescence programs in hematopoietic stem and progenitor cells (HSC/HSPCs) is perceived to be governed intrinsically and by microenvironmental co-adaptation. However, HSC programs dictating both transition and adaptability, remain poorly defined. Single cell multiome analysis divulging differential transcriptional activity between distinct HSPC states, indicated for the exclusive absence of Fli-1 motif from quiescent HSCs. We reveal that Fli-1 activity is essential for HSCs during regenerative hematopoiesis. Fli-1 directs activation programs while manipulating cellular sensory and output machineries, enabling HSPCs co-adoptability with a stimulated vascular niche. During regenerative conditions, Fli-1 presets and enables propagation of niche-derived Notch1 signaling. Constitutively induced Notch1 signaling is sufficient to recuperate functional HSC impairments in the absence of Fli-1. Applying FLI-1 modified-mRNA transduction into lethargic adult human mobilized HSPCs, enables their vigorous niche-mediated expansion along with superior engraftment capacities. Thus, decryption of stem cell activation programs offers valuable insights for immune regenerative medicine.

9.
Stem Cells ; 28(2): 308-17, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19967789

RESUMO

Hematopoietic stem cells (HSCs) can remain quiescent or they can enter the cell cycle, and either self-renew or differentiate. Although cyclin C and cyclin dependent kinase (cdk3) are essential for the transition from the G(0) to the G(1) phase of the cell cycle in human fibroblasts, the role of cyclin C in hematopoietic stem/progenitor cells (HSPCs) is not clear. We have identified an important role of cyclin C (CCNC) in regulating human HSPC quiescence, as knocking down CCNC expression in human cord blood CD34(+) cells resulted in a significant increase in quiescent cells that maintain CD34 expression. CCNC knockdown also promotes in vitro HSPC expansion and enhances their engraftment potential in sublethally irradiated immunodeficient mice. Our studies establish cyclin C as a critical regulator of the G(0)/G(1) transition of human HSPCs and suggest that modulating cyclin C levels may be useful for HSC expansion and more efficient engraftment.


Assuntos
Ciclina C/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Antígenos CD34/metabolismo , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiologia , Divisão Celular/genética , Divisão Celular/fisiologia , Células Cultivadas , Ciclina C/genética , Ciclina C/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia
10.
Sci Rep ; 11(1): 6104, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33731767

RESUMO

The human airway epithelium lining the bronchial tree contains basal cells that proliferate, differentiate, and communicate with other components of their microenvironment. One method that cells use for intercellular communication involves the secretion of exosomes and other extracellular vesicles (EVs). We isolated exosome-enriched EVs that were produced from an immortalized human airway basal cell line (BCi-NS1.1) and found that their secretion is increased by exposure to cigarette smoke extract, suggesting that this stress stimulates release of EVs which could affect signaling to other cells. We have previously shown that primary human airway basal cells secrete vascular endothelial growth factor A (VEGFA) which can activate MAPK signaling cascades in endothelial cells via VEGF receptor-2 (VEGFR2). Here, we show that exposure of endothelial cells to exosome-enriched airway basal cell EVs promotes the survival of these cells and that this effect also involves VEGFR2 activation and is, at least in part, mediated by VEGFA present in the EVs. These observations demonstrate that EVs are involved in the intercellular signaling between airway basal cells and the endothelium which we previously reported. The downstream signaling pathways involved may be distinct and specific to the EVs, however, as increased phosphorylation of Akt, STAT3, p44/42 MAPK, and p38 MAPK was not seen following exposure of endothelial cells to airway basal cell EVs.


Assuntos
Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Sistema de Sinalização das MAP Quinases , Produtos do Tabaco , Poluição por Fumaça de Tabaco , Linhagem Celular Transformada , Células Endoteliais/patologia , Vesículas Extracelulares/patologia , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Cancer Res ; 66(24): 11781-91, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17178874

RESUMO

The t(7;11)(p15;p15) translocation, observed in acute myelogenous leukemia and myelodysplastic syndrome, generates a chimeric gene where the 5' portion of the sequence encoding the human nucleoporin NUP98 protein is fused to the 3' region of HOXA9. Here, we show that retroviral-mediated enforced expression of the NUP98-HOXA9 fusion protein in cord blood-derived CD34(+) cells confers a proliferative advantage in both cytokine-stimulated suspension cultures and stromal coculture. This advantage is reflected in the selective expansion of hematopoietic stem cells as measured in vitro by cobblestone area-forming cell assays and in vivo by competitive repopulation of nonobese diabetic/severe combined immunodeficient mice. NUP98-HOXA9 expression inhibited erythroid progenitor differentiation and delayed neutrophil maturation in transduced progenitors but strongly enhanced their serial replating efficiency. Analysis of the transcriptosome of transduced cells revealed up-regulation of several homeobox genes of the A and B cluster as well as of Meis1 and Pim-1 and down-modulation of globin genes and of CAAT/enhancer binding protein alpha. The latter gene, when coexpressed with NUP98-HOXA9, reversed the enhanced proliferation of transduced CD34(+) cells. Unlike HOXA9, the NUP98-HOXA9 fusion was protected from ubiquitination mediated by Cullin-4A and subsequent proteasome-dependent degradation. The resulting protein stabilization may contribute to the leukemogenic activity of the fusion protein.


Assuntos
Antígenos CD34/fisiologia , Proteínas de Homeodomínio/genética , Proteínas Mutantes Quiméricas/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Células-Tronco/citologia , Antígenos CD/fisiologia , Divisão Celular , Linhagem Celular , Mapeamento Cromossômico , Cromossomos Humanos Par 11 , Cromossomos Humanos Par 7 , Clonagem Molecular , Ensaio de Unidades Formadoras de Colônias , Sangue Fetal , Fusão Gênica , Vetores Genéticos , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Translocação Genética , Cordão Umbilical
12.
Exp Hematol ; 34(11): 1505-16, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17046570

RESUMO

OBJECTIVE: The hematopoietic system is nurtured by a supportive stroma environment allowing maintenance and differentiation of hematopoietic stem cells (HSC). However, only a limited number of these stromal cell clones support hematopoiesis in the absence of cytokine supplementation. So far, only two bone marrow-derived stromal cell lines (OP9 and S17) are capable of inducing hematopoietic differentiation of totipotent murine and human embryonic stem cells (ESC). Here, the potential of more than 100 stromal cell lines developed from the aorta-gonado-mesonephros (AGM) region was investigated in supporting adult and embryonic hematopoiesis. In addition, extensive phenotypic analysis should elucidate possible mechanisms involved in maintenance of hematopoietic stem cell function. METHODS: More than 100 stromal cell clones derived from the AGM region of E10.5 mouse embryos were isolated. Hematopoietic stem cell support was tested for adult murine and human cord blood hematopoietic stem cells and hematopoietic cells derived from murine ESC. Genotypic and phenotypic characterization was performed including gene array analysis. RESULTS: It was demonstrated that multiple clones showed high efficiency in supporting maintenance and expansion of primitive murine and human hematopoietic progenitors. In addition, we demonstrated for the first time that AGM stromal cell lines are also potent inducers of hematopoietic differentiation of murine ESC. Microarray analysis of AGM lines revealed a characteristic genotype with expression of genes involved in regulating hematopoiesis as well as mesodermal and early B cell development. CONCLUSION: These AGM stromal cell lines may be of value in elucidating molecular mechanisms regulating early stem cell development and hematopoietic differentiation from ES-derived mesoderm.


Assuntos
Aorta/citologia , Gônadas/citologia , Hematopoese/fisiologia , Mesonefro/citologia , Células-Tronco/citologia , Células Estromais/citologia , Animais , Antígenos CD34/análise , Aorta/embriologia , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Linhagem Celular , Proliferação de Células , Clonagem Molecular , Sangue Fetal/citologia , Gônadas/embriologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/fisiologia , Células Estromais/fisiologia
13.
Methods Enzymol ; 418: 208-42, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17141038

RESUMO

Murine embryonic stem cells (mESC) readily form embryoid bodies (EBs) that exhibit hematopoietic differentiation. Methods based on EB formation or ESC coculture with murine bone marrow stromal cell lines have revealed pathways of both primitive and definitive hematopoietic differentiation progressing from primitive mesoderm via hemangioblasts to endothelium and hematopoietic stem and progenitor cells. The addition of specific hematopoietic growth factors and morphogens to these cultures enhances the generation of neutrophils, macrophages, megakaryocyte/platelets, and hemoglobinized mature red cells. In addition, selective culture systems have been developed to support differentiation into mature T lymphocytes, natural killer cells, B cells, and dendritic cells. In most cases, culture systems have been developed that support equivalent differentiation of various human ESC (hESC). The major obstacle to translation of ESC hematopoietic cultures to clinical relevance has been the general inability to produce hematopoietic stem cells (HSC) that can engraft adult, irradiated recipients. In this context, the pattern of ES hematopoietic development mirrors the yolk sac phase of hematopoiesis that precedes the appearance of engraftable HSC in the aorta-gonad-mesonephros region. Genetic manipulation of mESC hematopoietic progeny by upregulation of HOXB4 or STAT5 has led to greatly enhanced long- or short-term multilineage hematopoietic engraftment, suggesting that genetic or epigenetic manipulation of these pathways may lead to functional HSC generation from hESC.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Córion/citologia , Córion/fisiologia , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/fisiologia , Expressão Gênica , Hematopoese/genética , Humanos , Camundongos , Primatas , Células Estromais/citologia , Células Estromais/fisiologia
14.
Circ Res ; 94(6): 820-7, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14764454

RESUMO

Nuclear transfer techniques have been proposed as a strategy for generating an unlimited supply of rejuvenated and histocompatible stem cells for the treatment of cardiac diseases. For this purpose, c-kit-positive fetal liver stem cells obtained from cloned embryos were injected in the border zone of infarcted mice to induce tissue reconstitution. Cloned embryos were derived from somatic cell fusion between nuclei of cultured LacZ-positive fibroblasts and enucleated oocytes of a different mouse strain. We report that regenerating myocardium replaced 38% of the scar at 1 month. The rebuilt tissue expressed LacZ and was composed of myocytes and vessels connected with the coronary circulation. Myocytes were functionally competent and expressed contractile proteins, desmin, connexin43, and N-cadherin. These structural characteristics indicated that the new myocytes were electrically and mechanically coupled. Similarly, the formed coronary arterioles and capillary structures contained blood and contributed, therefore, to tissue oxygenation. Cardiac replacement resulted in an improvement of ventricular hemodynamics and in a reduction of diastolic wall stress. These beneficial effects were obtained by stem cell transdifferentiation and commitment to the cardiac cell lineages. Myocardial growth was independent from fusion of the injected stem cells with preexisting partner cells. In conclusion, c-kit-positive stem cells derived by nuclear transfer cloning restore infarcted myocardium. Although problems currently plague nuclear transplantation, including the potential for epigenetic and imprinting abnormalities, stem cells derived from cloned embryos are sufficiently normal to repair damaged tissue in vivo. Importantly, the magnitude of myocardial regeneration obtained in this study is significantly superior to that achieved with adult bone marrow cells.


Assuntos
Células Clonais/transplante , Transplante de Tecido Fetal , Coração/fisiologia , Infarto do Miocárdio/terapia , Técnicas de Transferência Nuclear , Transplante de Células-Tronco , Animais , Diferenciação Celular , Divisão Celular , Fusão Celular , Tamanho Celular , Clonagem de Organismos , Feminino , Fibroblastos/ultraestrutura , Genes Reporter , Injeções , Óperon Lac , Fígado/citologia , Fígado/embriologia , Masculino , Camundongos , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Neovascularização Fisiológica , Oócitos/ultraestrutura , Proteínas Proto-Oncogênicas c-kit/análise , Regeneração , Ultrassonografia
15.
Oncotarget ; 7(28): 43062-43075, 2016 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-27248664

RESUMO

DNA damaging agents cause rapid shrinkage of tumors and form the basis of chemotherapy for sarcomas despite significant toxicities. Drugs having superior efficacy and wider therapeutic windows are needed to improve patient outcomes. We used cell proliferation and apoptosis assays in sarcoma cell lines and benign cells; γ-H2AX expression, comet assay, immunoblot analyses and drug combination studies in vitro and in patient derived xenograft (PDX) models. BO-1055 caused apoptosis and cell death in a concentration and time dependent manner in sarcoma cell lines. BO-1055 had potent activity (submicromolar IC50) against Ewing sarcoma and rhabdomyosarcoma, intermediate activity in DSRCT (IC50 = 2-3µM) and very weak activity in osteosarcoma (IC50 >10µM) cell lines. BO-1055 exhibited a wide therapeutic window compared to other DNA damaging drugs. BO-1055 induced more DNA double strand breaks and γH2AX expression in cancer cells compared to benign cells. BO-1055 showed inhibition of tumor growth in A673 xenografts and caused tumor regression in cyclophosphamide resistant patient-derived Ewing sarcoma xenografts and A204 xenografts. Combination of BO-1055 and irinotecan demonstrated synergism in Ewing sarcoma PDX models. Potent activity on sarcoma cells and its relative lack of toxicity presents a strong rationale for further development of BO-1055 as a therapeutic agent.


Assuntos
Dano ao DNA , Compostos de Mostarda Nitrogenada/farmacologia , Compostos de Fenilureia/farmacologia , Sarcoma/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sinergismo Farmacológico , Células HCT116 , Humanos , Irinotecano , Células MCF-7 , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Compostos de Mostarda Nitrogenada/administração & dosagem , Compostos de Fenilureia/administração & dosagem , Sarcoma/genética , Sarcoma/patologia
16.
Radiat Res ; 185(1): 69-76, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26720799

RESUMO

It is not yet known whether hematopoietic stem and progenitor cells (HSPCs) are compromised in the aging population of atomic bomb (A-bomb) survivors after their exposure nearly 70 years ago. To address this, we evaluated age- and radiation-related changes in different subtypes of circulating HSPCs among the CD34-positive/lineage marker-negative (CD34(+)Lin(-)) cell population in 231 Hiroshima A-bomb survivors. We enumerated functional HSPC subtypes, including: cobblestone area-forming cells; long-term culture-initiating cells; erythroid burst-forming units; granulocyte and macrophage colony-forming units; and T-cell and natural killer cell progenitors using cell culture. We obtained the count of each HSPC subtype per unit volume of blood and the proportion of each HSPC subtype in CD34(+)Lin(-) cells to represent the lineage commitment trend. Multivariate analyses, using sex, age and radiation dose as variables, showed significantly decreased counts with age in the total CD34(+)Lin(-) cell population and all HSPC subtypes. As for the proportion, only T-cell progenitors decreased significantly with age, suggesting that the commitment to the T-cell lineage in HSPCs continuously declines with age throughout the lifetime. However, neither the CD34(+)Lin(-) cell population, nor HSPC subtypes showed significant radiation-induced dose-dependent changes in counts or proportions. Moreover, the correlations of the proportions among HSPC subtypes in the survivors properly revealed the hierarchy of lineage commitments. Taken together, our findings suggest that many years after exposure to radiation and with advancing age, the number and function of HSPCs in living survivors as a whole may have recovered to normal levels.


Assuntos
Células Sanguíneas/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos da radiação , Armas Nucleares/estatística & dados numéricos , Exposição à Radiação/estatística & dados numéricos , Sobreviventes/estatística & dados numéricos , Distribuição por Idade , Idoso , Idoso de 80 Anos ou mais , Células Sanguíneas/efeitos da radiação , Proliferação de Células/efeitos da radiação , Relação Dose-Resposta à Radiação , Feminino , Humanos , Japão/epidemiologia , Masculino , Distribuição por Sexo
17.
Artigo em Inglês | MEDLINE | ID: mdl-27169377

RESUMO

Accumulated DNA damage in hematopoietic stem cells is a primary mechanism of aging-associated dysfunction in human hematopoiesis. About 70 years ago, atomic-bomb (A-bomb) radiation induced DNA damage and functional decreases in the hematopoietic system of A-bomb survivors in a radiation dose-dependent manner. The peripheral blood cell populations then recovered to a normal range, but accompanying cells derived from hematopoietic stem cells still remain that bear molecular changes possibly caused by past radiation exposure and aging. In the present study, we evaluated radiation-related changes in the frequency of phosphorylated (Ser-139) H2AX (γH2AX) foci formation in circulating CD34-positive/lineage marker-negative (CD34+Lin-) hematopoietic stem and progenitor cells (HSPCs) among 226Hiroshima A-bomb survivors. An association between the frequency of γH2AX foci formation in HSPCs and the radiation dose was observed, but the γH2AX foci frequency was not significantly elevated by past radiation. We found a negative correlation between the frequency of γH2AX foci formation and the length of granulocyte telomeres. A negative interaction effect between the radiation dose and the frequency of γH2AX foci was suggested in a proportion of a subset of HSPCs as assessed by the cobblestone area-forming cell assay (CAFC), indicating that the self-renewability of HSPCs may decrease in survivors who were exposed to a higher radiation dose and who had more DNA damage in their HSPCs. Thus, although many years after radiation exposure and with advancing age, the effect of DNA damage on the self-renewability of HSPCs may be modified by A-bomb radiation exposure.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco/citologia , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Dano ao DNA/genética , Dano ao DNA/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Pessoa de Meia-Idade , Células-Tronco/metabolismo
18.
Stem Cells Dev ; 14(5): 505-16, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16305336

RESUMO

OP-9 cells are stromal cells derived from macrophage colony-stimulating factor (M-CSF)-deficient osteopetrotic mice. To evaluate the OP-9 capability to sustain long-term hematopoiesis, we reported the expansion of granulocyte colony-stimulating factor (G-CSF)-mobilized human peripheral blood (PB) CD34(+) cells in co-culture with murine OP-9 and MS-5 stromal cells, either transfected with various combinations of adenovectors (Ad) expressing c-kit ligand (KL) (either soluble or transmembrane form), thrombopoietin (TPO), flt-3/flk2 ligand (FL), and granulocyte-macrophage (GM)-CSF or with weekly addition of these cytokines. Expression of TPO as well as association of TPO, FL, and KL increased progenitor cell and week-6 cobblestone area forming cell (CAFC) production in all stromal co-cultures. Similar progenitor expansion was obtained by weekly addition of soluble cytokine. Five weeks of co-culture with OP9 and TPO, FL + KL resulted in the greatest expansion of progenitor cells and week-6 CAFC as measured by secondary assay on MS-5. In contrast to MS-5 and TPO or TPO + FL + KL cultures where hematopoiesis declined by week 4, progenitor as well as week-6 CAFC expansion continued for over 3 months in TPO + FL + KL OP9 cocultures. This was associated with decrease of CD14(+) macrophage production. The addition of human macrophage (M)-CSF or CD14(+) cells to the co-culture decrease progenitor and stem cell expansion; however, murine M-CSF to OP-9 co-cultures did not decrease progenitor expansion. High levels of stromal-derived factor-1 (SDF-1) production by MS-5 and low or absent production by OP-9 may account for stem cell adhesion and CAFC formation in the former cultures and the predominance of stem and progenitor cells in the nonadherent fraction in the latter cultures.


Assuntos
Antígenos CD34/metabolismo , Hematopoese/fisiologia , Proteínas de Membrana/metabolismo , Osteopetrose , Fator de Células-Tronco/metabolismo , Células Estromais/metabolismo , Trombopoetina/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Adjuvantes Imunológicos/metabolismo , Animais , Antígenos CD34/genética , Técnicas de Cultura de Células/métodos , Linhagem Celular , Quimiotaxia/fisiologia , Técnicas de Cocultura , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteínas de Membrana/genética , Camundongos , Fator de Células-Tronco/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Células Estromais/citologia , Trombopoetina/genética
19.
Clin Cancer Res ; 10(21): 7207-19, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15534094

RESUMO

The Wilms tumor protein (WT1) is overexpressed in most acute and chronic leukemias. To develop a practicable, clinically applicable approach for generation of WT1-specific T cells and to comparatively evaluate the immunogenicity of WT1 in normal individuals, we sensitized T cells from 13 HLA-A0201+ and 5 HLA-A2402+ donors with autologous EBV-transformed B cells or cytokine-activated monocytes, loaded with the HLA-A0201-binding WT1 peptides (126-134)RMFPNAPYL or (187-195)SLGEQQYSV or a newly identified HLA-A2402-binding WT1 peptide (301-310)RVPGVAPTL. WT1-specific T cells were regularly generated from each donor. T cells sensitized with peptide-loaded EBV-transformed B cells generated higher numbers of WT1-specific T cells than peptide-loaded cytokine-activated monocytes. Contrary to expectations, the frequencies of WT1 peptide-specific T cells were equivalent to those generated against individual highly immunogenic HLA-A0201-binding EBV peptides. Each of these T-cell lines specifically killed WT1+ leukemias and solid tumors in an HLA-restricted manner but did not lyse autologous or HLA-matched normal CD34+ hematopoietic progenitor cells or reduce their yield of colony-forming unit-granulocyte-macrophage (CFU-GM), burst-forming unit erythroid (BFU-E), or mixed colonies (CFU-mix). Furthermore, WT1 peptide-specific T cells after adoptive transfer into nonobese diabetic-severe combined immunodeficient mice bearing subcutaneous xenografts of WT1+ and WT1- HLA-A0201+ leukemias preferentially accumulated in and induced regressions of WT1+ leukemias that expressed the restricting HLA allele. Such cells are clinically applicable and may prove useful for adoptive cell therapy of WT1+ malignant diseases in humans.


Assuntos
Herpesvirus Humano 4/genética , Linfócitos T/imunologia , Proteínas WT1/farmacologia , Alelos , Animais , Antígenos CD34/biossíntese , Linhagem Celular Tumoral , Movimento Celular , Citocinas/metabolismo , Genes do Tumor de Wilms , Humanos , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos SCID , Monócitos/metabolismo , Transplante de Neoplasias , Peptídeos/química , Ligação Proteica , Sensibilidade e Especificidade , Linfócitos T/metabolismo , Fatores de Tempo , Proteínas WT1/química
20.
Exp Hematol ; 32(3): 300-7, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15003316

RESUMO

OBJECTIVE: Stromal-derived factor-1 (SDF-1/CXCL12) is chemotactic for lympho/hematopoietic stem cells. We have previously shown that increasing peripheral blood (PB) levels of SDF-1 with adenovectors expressing human SDF-1 complementary DNA (ad-SDF-1) leads to hematopoietic stem cell mobilization as well as migration of megakaryocytes and thrombocytosis in mice. Herein, we studied the in vivo effects of ad-SDF-1 and of an analogue peptide of SDF-1 (CTCE-0214) on human hematopoiesis in a xenotransplant model. MATERIALS AND METHODS: Sublethally irradiated (300 cGY) NOD/SCID mice transplanted with human cord blood mononuclear cells (CB MNC) were injected with ad-SDF-1 (10(9) plaque forming units, i.v., x 1) or CTCE-0214 (10 mg/kg/dose, i.v. q 24 hours x 7). Effects on megakaryocytopoiesis (CD41+ cells and platelets) as well as stem cell mobilization were monitored. RESULTS: CB MNC in NOD/SCID mice are able to differentiate into CD41+ cells and platelets, peaking at week 9 at a mean of 3.7 x 10(3)/microL. i.v. injection of ad-SDF-1 increased human CD41+ cells by day 4 in PB and was followed by an increase in human platelet production by day 5, with return to baseline by day 30. Human colony-forming cells (CFC) were mobilized from bone marrow to spleen (by day 6-13) and to PB (by day 13). Human CD34+ and CD33+ cells were mobilized by this treatment as well. A novel SDF-1 peptide agonist (CTCE-0214) also mobilized human CFC and enhanced human thrombopoiesis. CONCLUSION: SDF-1 and its analogue may be of clinical value in stimulating platelet recovery after chemo/radiation treatment as well as in stem cell mobilization.


Assuntos
Plaquetas/citologia , Quimiocinas CXC/farmacologia , Hematopoese/efeitos dos fármacos , Mobilização de Células-Tronco Hematopoéticas/métodos , Megacariócitos/efeitos dos fármacos , Adenoviridae/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Quimiocina CXCL12 , Quimiocinas CXC/agonistas , Quimiocinas CXC/sangue , Quimiocinas CXC/genética , Avaliação Pré-Clínica de Medicamentos , Sangue Fetal/citologia , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Quimera por Radiação , Proteínas Recombinantes de Fusão/fisiologia , Trombopoetina/genética , Trombopoetina/farmacologia , Transdução Genética , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA