Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Circulation ; 135(25): 2505-2523, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28381471

RESUMO

BACKGROUND: The mechanisms underlying the dedifferentiation and lineage conversion of adult human fibroblasts into functional endothelial cells have not yet been fully defined. Furthermore, it is not known whether fibroblast dedifferentiation recapitulates the generation of multipotent progenitors during embryonic development, which give rise to endothelial and hematopoietic cell lineages. Here we established the role of the developmental transcription factor SOX17 in regulating the bilineage conversion of fibroblasts by the generation of intermediate progenitors. METHODS: CD34+ progenitors were generated after the dedifferentiation of human adult dermal fibroblasts by overexpression of pluripotency transcription factors. Sorted CD34+ cells were transdifferentiated into induced endothelial cells and induced erythroblasts using lineage-specific growth factors. The therapeutic potential of the generated cells was assessed in an experimental model of myocardial infarction. RESULTS: Induced endothelial cells expressed specific endothelial cell surface markers and also exhibited the capacity for cell proliferation and neovascularization. Induced erythroblasts expressed erythroid surface markers and formed erythroid colonies. Endothelial lineage conversion was dependent on the upregulation of the developmental transcription factor SOX17, whereas suppression of SOX17 instead directed the cells toward an erythroid fate. Implantation of these human bipotential CD34+ progenitors into nonobese diabetic/severe combined immunodeficiency (NOD-SCID) mice resulted in the formation of microvessels derived from human fibroblasts perfused with mouse and human erythrocytes. Endothelial cells generated from human fibroblasts also showed upregulation of telomerase. Cell implantation markedly improved vascularity and cardiac function after myocardial infarction without any evidence of teratoma formation. CONCLUSIONS: Dedifferentiation of fibroblasts to intermediate CD34+ progenitors gives rise to endothelial cells and erythroblasts in a SOX17-dependent manner. These findings identify the intermediate CD34+ progenitor state as a critical bifurcation point, which can be tuned to generate functional blood vessels or erythrocytes and salvage ischemic tissue.


Assuntos
Antígenos CD34/fisiologia , Desdiferenciação Celular/fisiologia , Células Endoteliais/fisiologia , Eritroblastos/fisiologia , Fibroblastos/fisiologia , Fatores de Transcrição SOXF/fisiologia , Células-Tronco/fisiologia , Animais , Células Cultivadas , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
2.
Ann Hematol ; 95(2): 173-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26555286

RESUMO

The nonhematopoietic bone marrow (BM) microenvironment provides a functional niche for hematopoietic cell maintenance, recruitment, and differentiation. It consists of multiple cell types including vasculature, bone, adipose tissue, and fibroblast-like bone marrow stromal cells (BMSC), which can be summarized under the generic term niche cells. BMSC express Toll-like receptors (TLRs) and are capable to respond to TLR-agonists by changing their cytokine expression pattern in order to more efficiently support hematopoiesis. Here, we show that in addition to enhanced myeloid colony formation from human CD34+ cells, lipopolysaccharide (LPS) stimulation retains overall higher numbers of CD34+ cells in co-culture assays using BMSC, with eightfold more CD34+ cells that underwent up to three divisions as compared to non-stimulated assays. When subjected to cytokine-supplemented myeloid colony-forming unit (CFU) assays or transplanted into newborn RAG2(-/-) γc (-/-) mice, CD34(+) cells from LPS-stimulated BMSC cultures give rise to the full spectrum of myeloid colonies and T and B cells, respectively, thus supporting maintenance of myeloid and lymphoid primed hematopoietic progenitor cells (HPCs) under inflammatory conditions. Collectively, we suggest that BMSC enhance hematopoiesis during inflammatory conditions to support the replenishment of innate immune effector cells and to prevent the exhaustion of the hematopoietic stem and progenitor cell (HSPC) pool.


Assuntos
Diferenciação Celular/fisiologia , Lipopolissacarídeos/farmacologia , Células-Tronco Mesenquimais/fisiologia , Células Mieloides/fisiologia , Animais , Antígenos CD34/fisiologia , Transplante de Medula Óssea/métodos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Mieloides/efeitos dos fármacos
3.
Biol Reprod ; 93(2): 37, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26108791

RESUMO

Repeated and dramatic pregnancy-induced uterine enlargement and remodeling throughout reproductive life suggests the existence of uterine smooth muscle stem/progenitor cells. The aim of this study was to isolate and characterize stem/progenitor-like cells from human myometrium through identification of specific surface markers. We here identify CD49f and CD34 as markers to permit selection of the stem/progenitor cell-like population from human myometrium and show that human CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells exhibit stem cell-like properties. These include side population phenotypes, an undifferentiated status, high colony-forming ability, multilineage differentiation into smooth muscle cells, osteoblasts, adipocytes, and chondrocytes, and in vivo myometrial tissue reconstitution following xenotransplantation. Furthermore, CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells proliferate under hypoxic conditions in vitro and, compared with the untreated nonpregnant myometrium, show greater expansion in the estrogen-treated nonpregnant myometrium and further in the pregnant myometrium in mice upon xenotransplantation. These results suggest that the newly identified myometrial stem/progenitor-like cells influenced by hypoxia and sex steroids may participate in pregnancy-induced uterine enlargement and remodeling, providing novel insights into human myometrial physiology.


Assuntos
Antígenos CD34/genética , Antígenos CD34/fisiologia , Integrina alfa6/genética , Integrina alfa6/fisiologia , Miométrio/metabolismo , Células-Tronco/fisiologia , Útero/fisiologia , Animais , Diferenciação Celular , Hipóxia Celular , Linhagem da Célula/genética , Feminino , Glicoforinas/biossíntese , Glicoforinas/genética , Células-Tronco Hematopoéticas , Humanos , Camundongos , Miométrio/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Gravidez
4.
Exp Dermatol ; 23(6): 407-13, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24750467

RESUMO

Hair follicles have characteristic sizes corresponding to their cycle-specific stage. However, how the anagen hair follicle specifies its size remains elusive. Here, we showed that in response to prolonged ectopic Wnt10b-mediated ß-catenin activation, regenerating anagen hair follicles grew larger in size. In particular, the hair bulb, dermal papilla and hair shaft became enlarged, while the formation of different hair types (Guard, Awl, Auchene and Zigzag) was unaffected. Interestingly, we found that the effect of exogenous WNT10b was mainly on Zigzag and less on the other kinds of hairs. We observed dramatically enhanced proliferation within the matrix, DP and hair shaft of the enlarged AdWnt10b-treated hair follicles compared with those of normal hair follicles at P98. Furthermore, expression of CD34, a specific hair stem cell marker, was increased in its number to the bulge region after AdWnt10b treatment. Ectopic expression of CD34 throughout the ORS region was also observed. Many CD34-positive hair stem cells were actively proliferating in AdWnt10b-induced hair follicles. Importantly, subsequent co-treatment with the Wnt inhibitor, DKK1, reduced hair follicle enlargement and decreased proliferation and ectopic localization of hair stem cells. Moreover, injection of DKK1 during early anagen significantly reduced the width of prospective hairs. Together, these findings strongly suggest that Wnt10b/DKK1 can modulate hair follicle size during hair regeneration.


Assuntos
Folículo Piloso/anatomia & histologia , Cabelo/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Regeneração/fisiologia , Proteínas Wnt/fisiologia , Animais , Antígenos CD34/fisiologia , Proliferação de Células/fisiologia , Feminino , Folículo Piloso/citologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Plasmídeos , Transdução de Sinais/fisiologia , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/genética , beta Catenina/fisiologia
6.
Haematologica ; 98(12): 1964-71, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24038022

RESUMO

Patients who have undergone autologous stem cell transplantation are subsequently more susceptible to chemotherapy-induced bone marrow toxicity. In the present study, bone marrow primitive progenitor cells were examined one year after autologous stem cell transplantation and compared with normal bone marrow and mobilized peripheral blood stem cells. Post-transplantation bone marrow contained a significantly lower percentage of quiescent cells in the CD34(+)/CD38(low) fraction compared to normal bone marrow. In addition, we observed a strong decrease in stem cell/primitive progenitor frequency in post-transplantation CD34(+) cells as defined by long-term culture assays. Measurement of the levels of reactive oxygen species by flow cytometry revealed comparable levels in post-transplantation and normal bone marrow CD34(+)/CD38(low) cells, while significantly higher levels of reactive oxygen species were observed in CD34(+)/CD38(high) cells following autologous stem cell transplantation compared to normal bone marrow. Moreover, post-transplantation CD34(+) bone marrow cells demonstrated an increased sensitivity to buthionine sulfoximine, a trigger for endogenous production of reactive oxygen species. Gene expression analysis on CD34(+) cells revealed a set of 195 genes, including HMOX1, EGR1, FOS and SIRPA that are persistently down-regulated in mobilized peripheral blood cells and post-transplantation bone marrow compared to normal bone marrow. In conclusion, our data indicate that the diminished regenerative capacity of bone marrow following autologous stem cell transplantation is possibly related to a loss of quiescence and a reduced tolerability to oxidative stress.


Assuntos
ADP-Ribosil Ciclase 1/fisiologia , Antígenos CD34/fisiologia , Transplante de Células-Tronco Hematopoéticas/tendências , Adulto , Idoso , Células Cultivadas , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Transplante Autólogo/tendências
7.
Vet Surg ; 42(2): 137-46, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23373667

RESUMO

OBJECTIVE: To identify the optimum intra-articular multipotent stromal cell (MSC) tissue source in the canine stifle. STUDY DESIGN: Experimental. SAMPLE POPULATION: Infrapatellar adipose tissue, synovium lining the joint capsule, and synovium surrounding the cranial cruciate ligament (CrCL) from normal stifles of 6 dogs. METHODS: Nucleated cell density for each tissue was determined, and cell doublings (CD) and doubling times (DT) were quantified for expansion rates. Adipogenic, osteogenic, and chondrogenic differentiation was confirmed with light microscopy. Fibroblastic, adipogenic, and osteogenic colony forming unit frequencies were determined for multipotentiality. Tissue-specific target gene expression was assessed, and percentages of CD29(+) , CD34(+) , CD44(+) , CD45(+) , and CD90(+) cells quantified. RESULTS: Adipose tissue had the highest MSC density (ASC). The CD decreased with increasing passages for all cell types, and ASC values tended to be higher. Multipotentiality decreased with passage, but remained highest in ASC. Tissue-specific target gene expression was higher in induced versus noninduced cells, and ASCs had the highest upregulation across passages. Most cells were CD29(+) , CD44(+) , CD90(+) , and percentages decreased with passage. Within cell types, there were more CD29(+) ASC in early passages and more CD44(+) and CD90(+) ASC in later passages. CONCLUSIONS: ASC had the highest in vitro expansion rates, CFU frequencies, tissue-specific target gene expression, and percentages of MSC immunophenotypes.


Assuntos
Tecido Adiposo/citologia , Antígenos CD/fisiologia , Células-Tronco Multipotentes/fisiologia , Células Estromais/citologia , Animais , Ligamento Cruzado Anterior/citologia , Antígenos CD/imunologia , Antígenos CD34/fisiologia , Contagem de Células/veterinária , Divisão Celular/fisiologia , Cães , Feminino , Receptores de Hialuronatos/fisiologia , Imunofenotipagem/veterinária , Integrina beta1/fisiologia , Antígenos Comuns de Leucócito/fisiologia , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/imunologia , Joelho de Quadrúpedes/citologia , Células Estromais/imunologia , Células Estromais/fisiologia , Antígenos Thy-1/fisiologia
8.
J Immunol ; 184(3): 1292-9, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20038636

RESUMO

CD34 is a cell surface sialomucin expressed by hematopoietic precursors, eosinophils, mast cells, and vascular endothelia and is suggested to play an integral role in mucosal inflammatory responses. Although Cd34(-/-) mice have normal hematopoietic cell subsets in peripheral tissues at steady state, they exhibit a cell recruitment defect when challenged, offering a unique opportunity to distinguish between local inflammatory cell proliferation and peripheral recruitment in disease. Autoimmune arthritis is an inflammatory disease dependent on hematopoietic infiltration, and in this study, we have examined the role of CD34 in disease development and progression. Using an autoimmune serum transfer model, arthritis was induced in C57BL/6 wild-type and Cd34(-/-) mice. Surprisingly, we found that Cd34(-/-) mice were more susceptible to arthritis than wild-type mice. We examined mast cell-transplanted, eosinophil-deficient, and bone marrow-chimeric mice to determine the role of CD34 expression on disease progression. These experiments excluded CD34-deficient mast cells, eosinophils, or hematopoietic cells as the cause of the exacerbated disease. Further study demonstrated that Cd34(-/-) mice exhibit increased vascular leakage at onset of disease and in response to TNF, which correlated with a subsequent increase in disease severity. We conclude that loss of CD34 expression leads to increased vascular permeability in the joints at onset of disease, leading to exacerbated arthritic disease in Cd34(-/-) mice.


Assuntos
Antígenos CD34/genética , Artrite Experimental/genética , Artrite Experimental/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Permeabilidade Capilar/genética , Permeabilidade Capilar/imunologia , Animais , Antígenos CD34/fisiologia , Artrite Experimental/patologia , Artrite Experimental/fisiopatologia , Doenças Autoimunes/patologia , Doenças Autoimunes/fisiopatologia , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/patologia , Células Cultivadas , Progressão da Doença , Imunofenotipagem , Masculino , Mastócitos/imunologia , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Índice de Gravidade de Doença
9.
Eur Heart J ; 32(10): 1197-206, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21362705

RESUMO

In the absence of effective endogenous repair mechanisms after cardiac injury, cell-based therapies have rapidly emerged as a potential novel therapeutic approach in ischaemic heart disease. After the initial characterization of putative endothelial progenitor cells and their potential to promote cardiac neovascularization and to attenuate ischaemic injury, a decade of intense research has examined several novel approaches to promote cardiac repair in adult life. A variety of adult stem and progenitor cells from different sources have been examined for their potential to promote cardiac repair and regeneration. Although early, small-scale clinical studies underscored the potential effects of cell-based therapy largely by using bone marrow (BM)-derived cells, subsequent randomized-controlled trials have revealed mixed results that might relate, at least in part, to differences in study design and techniques, e.g. differences in patient population, cell sources and preparation, and endpoint selection. Recent meta-analyses have supported the notion that administration of BM-derived cells may improve cardiac function on top of standard therapy. At this stage, further optimization of cell-based therapy is urgently needed, and finally, large-scale clinical trials are required to eventually proof its clinical efficacy with respect to outcomes, i.e. morbidity and mortality. Despite all promises, pending uncertainties and practical limitations attenuate the therapeutic use of stem/progenitor cells for ischaemic heart disease. To advance the field forward, several important aspects need to be addressed in carefully designed studies: comparative studies may allow to discriminate superior cell populations, timing, dosing, priming of cells, and delivery mode for different applications. In order to predict benefit, influencing factors need to be identified with the aim to focus resources and efforts. Local retention and fate of cells in the therapeutic target zone must be improved. Further understanding of regenerative mechanisms will enable optimization at all levels. In this context, cell priming, bionanotechnology, and tissue engineering are emerging tools and may merge into a combined biological approach of ischaemic tissue repair.


Assuntos
Isquemia Miocárdica/terapia , Transplante de Células-Tronco/métodos , Antígeno AC133 , Doença Aguda , Antígenos CD/fisiologia , Antígenos CD34/fisiologia , Doença Crônica , Células-Tronco Embrionárias/transplante , Previsões , Glicoproteínas/fisiologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/terapia , Peptídeos/fisiologia , Células-Tronco Pluripotentes/transplante
10.
Blood ; 113(11): 2470-7, 2009 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-18840707

RESUMO

Interleukin-15 (IL-15) is essential for natural killer (NK) cell differentiation. In this study, we assessed whether the receptor tyrosine kinase Axl and its ligand, Gas6, are involved in IL-15-mediated human NK differentiation from CD34(+) hematopoietic progenitor cells (HPCs). Blocking the Axl-Gas6 interaction with a soluble Axl fusion protein (Axl-Fc) or the vitamin K inhibitor warfarin significantly diminished the absolute number and percentage of CD3(-)CD56(+) NK cells derived from human CD34(+) HPCs cultured in the presence of IL-15, probably resulting in part from reduced phosphorylation of STAT5. In addition, CD3(-)CD56(+) NK cells derived from culture of CD34(+) HPCs with IL-15 and Axl-Fc had a significantly diminished capacity to express interferon-gamma or its master regulator, T-BET. Culture of CD34(+) HPCs in the presence of c-Kit ligand and Axl-Fc resulted in a significant decrease in the frequency of NK precursor cells responding to IL-15, probably the result of reduced c-Kit phosphorylation. Collectively, our data suggest that the Axl/Gas6 pathway contributes to normal human NK-cell development, at least in part via its regulatory effects on both the IL-15 and c-Kit signaling pathways in CD34(+) HPCs, and to functional NK-cell maturation via an effect on the master regulatory transcription factor T-BET.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Interleucina-15/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Proteínas Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Antígenos CD34/metabolismo , Antígenos CD34/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Citocinas/farmacologia , Citocinas/fisiologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interleucina-15/fisiologia , Células K562 , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/fisiologia , Proteínas Oncogênicas/genética , Proteínas Proto-Oncogênicas , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas c-kit/fisiologia , Receptores Proteína Tirosina Quinases/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas com Domínio T/fisiologia , Receptor Tirosina Quinase Axl
11.
J Immunol ; 182(9): 5507-14, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380799

RESUMO

Reports showing that W/W(v) mice are protected from experimental autoimmune encephalomyelitis (EAE, a murine model of multiple sclerosis), have implicated mast cells as an essential component in disease susceptibility, but the role of mast cell trafficking has not been addressed. In this study, we have used both mast cell transplantation and genetic mutations (Cd34(-/-), W/W(v), W(sh)/W(sh)) to investigate the role of mast cell trafficking in EAE in detail. We show, for the first time, that bone marrow-derived mast cells are actively recruited to the CNS during EAE. Unexpectedly, however, we found that EAE develops unabated in two independent genetic backgrounds in the complete absence of mast cells or bone marrow-derived mast cell reconstitution. We conclude that although mast cells do accumulate in the brain and CNS during demyelinating disease via peripheral mast cell trafficking, they are completely dispensable for development of disease.


Assuntos
Movimento Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Predisposição Genética para Doença , Mastócitos/imunologia , Mastócitos/patologia , Medula Espinal/imunologia , Medula Espinal/patologia , Transferência Adotiva , Sequência de Aminoácidos , Animais , Antígenos CD34/genética , Antígenos CD34/fisiologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Movimento Celular/genética , Encefalomielite Autoimune Experimental/genética , Feminino , Mastócitos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Dados de Sequência Molecular , Medula Espinal/metabolismo
12.
Mol Ther ; 18(2): 404-12, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19861952

RESUMO

Cells localized in the bronchioalveolar duct junction of the murine lung have been identified as potential bronchioalveolar stem cells. Based on the surface marker expression, two main phenotypes have been proposed: Sca-1(+), CD34(+), CD45(-), Pecam(-) and Sca-1(low), CD34(-) CD45(-), Pecam(-) cells. An increase in the number of Sca-1(+), CD34(+) CD45(-), Pecam(-) cells and activation of the sonic hedgehog (Shh) pathway was observed following unilateral pneumonectomy and naphthalene-induced airway injury. Overexpression of Shh in the respiratory tract also resulted in an increase of this cell population. Syngeneic transplantation of beta-galactosidase-expressing bone marrow cells demonstrated that the increase of Sca-1(+), CD34(+), CD45(-), Pecam(-) cells in the lung was a result of local proliferation. Intratracheal administration of purified Shh-stimulated Sca-1(+), CD45(-), Pecam(-) cells coexpressing CD34 to syngeneic mice following pneumonectomy resulted in engraftment of these cells predominantly in the airways for up to 3 months, whereas Sca-1(-), CD45(-), Pecam(-) cells did not engraft. This study suggests that local Sca-1(+), CD34(+), CD45(-), Pecam(-) cells are stimulated during compensatory lung growth, following airway injury and overexpression of Shh and have some potential to engraft in the airways, without showing clonal properties in vivo.


Assuntos
Antígenos CD34/fisiologia , Antígenos Ly/fisiologia , Proteínas Hedgehog/fisiologia , Lesão Pulmonar/metabolismo , Proteínas de Membrana/fisiologia , Adenoviridae/genética , Animais , Antígenos CD34/genética , Antígenos Ly/genética , Células Cultivadas , Feminino , Citometria de Fluxo , Imunofluorescência , Proteínas Hedgehog/genética , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/fisiologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Naftalenos/toxicidade , Pneumonectomia
13.
Am J Physiol Endocrinol Metab ; 299(1): E110-6, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20424141

RESUMO

Endothelial progenitor cells (EPCs) play an important role in angiogenesis, which is essential for numerous physiological processes as well as tumor growth. Several microRNAs (miRNAs) have been reported to be involved in angiogenesis. MiR-34a, recently reported as a tumor suppressor, has been found to target silent information regulator 1 (Sirt1), leading to cell cycle arrest or apoptosis. However, the role of miR-34a in EPC-mediated angiogenesis was unknown. The present study tested the hypothesis that miR-34a inhibits EPC-mediated angiogenesis by inducing senescence via suppressing Sirt1. Bone marrow-derived EPCs from adult male Sprague-Dawley rats were used. Results of flow cytometry showed that EPCs after 7 days of culture expressed both stem cell markers CD34 and CD133 and endothelial cell markers VEGFR-2 (flk-1) and VE-cadherin. MiR-34a was expressed in normal EPCs, and overexpression of miR-34a via its mimic transfection significantly increased its expression and impaired in vitro EPC angiogenesis. MiR-34a overexpression led to a significantly increased EPC senescence, paralleled with an approximately 40% Sirt1 reduction. Furthermore, knockdown of Sirt1 by its siRNA resulted in diminished EPC angiogenesis and increased senescence. Finally, overexpression of miR-34a increased the level of Sirt1 effector-acetylated forkhead box O transcription factors 1 (FoxO1), an effect mimicked in EPCs following Sirt1 knockdown. In conclusion, miR-34a impairs EPC-mediated angiogenesis by induction of senescence via inhibiting Sirt1.


Assuntos
Senescência Celular/fisiologia , MicroRNAs/fisiologia , Neovascularização Fisiológica/fisiologia , Sirtuína 1/fisiologia , Células-Tronco/fisiologia , Antígeno AC133 , Animais , Antígenos CD/análise , Antígenos CD/fisiologia , Antígenos CD34/análise , Antígenos CD34/fisiologia , Western Blotting , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/fisiologia , Regulação da Expressão Gênica , Glicoproteínas/análise , Glicoproteínas/fisiologia , Masculino , MicroRNAs/genética , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/fisiologia , Peptídeos/análise , Peptídeos/fisiologia , RNA/química , RNA/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/antagonistas & inibidores , Células-Tronco/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
14.
J Exp Med ; 183(3): 837-45, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8642288

RESUMO

Erythropoietin (EPO) is the primary humoral regulator of erythropoiesis and no other factor has previously been reported to support proliferation and terminal maturation of erythroid cells from hemopoietic stem cells. Here we show that stimulation of glycoprotein (gp130) by a combination of recombinant human soluble interleukin 6 receptor (sIL-6R) and IL-6 but not sIL-6R or IL-6 alone can support proliferation, differentiation, and terminal maturation of erythroid cells in the absence of EPO from purified human CD34+ cells in suspension culture containing stem cell factor (SCF). A number of erythroid bursts and mixed erythroid colonies also developed in methylcellulose culture under the same combination. The addition of anti-gp130 monoclonal antibodies but not anti-EPO antibody to the same culture completely abrogated the generation of erythroid cells. These results clearly demonstrate that mature erythroid cells can be emerged from hemopoietic progenitors without EPO in vitro. Together with the previous reports that human sera contain detectable levels of sIL-6R, IL-6, and SCF, current data suggest that gp130 signaling in association with c-kit activation may play a role in human erythropoiesis in vivo.


Assuntos
Antígenos CD34/análise , Antígenos CD/fisiologia , Eritrócitos/citologia , Eritropoese , Eritropoetina/fisiologia , Células-Tronco Hematopoéticas/citologia , Glicoproteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas c-kit/fisiologia , Receptores de Interleucina/fisiologia , Transdução de Sinais , Antígenos CD/análise , Antígenos CD/biossíntese , Antígenos CD34/fisiologia , Diferenciação Celular/efeitos dos fármacos , Divisão Celular , Células Clonais , Receptor gp130 de Citocina , Eritropoetina/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interleucina-6/farmacologia , Cinética , Receptores de Interleucina/biossíntese , Receptores de Interleucina-6 , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/farmacologia
15.
Cancer Sci ; 101(3): 631-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20028384

RESUMO

Stem cells of acute myeloid leukemia (AML) have been identified as immunodeficient mouse-repopulating cells with a Lin(-)CD34(+)38(-) phenotype similar to normal hematopoietic stem cells. To identify the leukemia-propagating stem cell fraction of Philadelphia chromosome-positive (Ph(+)) leukemia, we serially transplanted human leukemia cells from patients with chronic myeloid leukemia blast crisis (n = 3) or Ph(+) acute lymphoblastic leukemia (n = 3) into NOD/SCID/IL-2Rgammac(-/-) mice. Engrafted cells were almost identical to the original leukemia cells as to phenotypes, IGH rearrangements, and karyotypes. CD34(+)CD38(-)CD19(+), CD34(+)38(+)CD19(+), and CD34(-)CD38(+)CD19(+) fractions could self-renew and transfer the leukemia, whereas the CD34(-)CD38(+)CD19(+) fraction did not stably propagate in NOD/SCID mice. These findings suggest that leukemia-repopulating cells in transformed Ph(+) leukemia are included in a lineage-committed but multilayered fraction, and that CD34(+) leukemia cells potentially emerge from CD34(-) populations.


Assuntos
Antígenos CD34/fisiologia , Linhagem da Célula , Leucemia/patologia , Cromossomo Filadélfia , Receptores de Interleucina-2/fisiologia , ADP-Ribosil Ciclase 1/análise , Animais , Antígenos CD34/análise , Humanos , Leucemia/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
16.
Cytokine ; 52(3): 245-51, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20829062

RESUMO

Inflammatory responses have been implicated in the elaboration of several forms of central nervous system injury, including cerebral vasospasm after subarachnoid hemorrhage (SAH). A critical event participating in such responses is the recruitment of circulating leukocytes into the inflammatory site. CD34 is a key adhesion molecule responsible for recruitment of monocytes/macrophages and the attachment of leukocytes to endothelial cells. However, it has not been investigated whether, and to what degree, CD34 is induced by SAH and also the role of CD34 in the pathogenesis of cerebral vasospasm following SAH remains unknown. Experiment 1 aimed to investigate the timecourse of the CD34 expression in the basilar artery after SAH. In experiment 2, we chose the maximum time point of vasospasm (day 3) and assessed the effect of monoclonal antibody against CD34 on regulation of cerebral vasospasm. As a result, the elevated expression of CD34 was detected in the basilar artery after SAH and peaked on day 3. After intracisternal administration of CD34 monoclonal antibody, the vasospasm was markedly attenuated after blood injection on day 3. Our results suggest that CD34 is increasingly expressed in a parallel time course to the development of cerebral vasospasm in a rat experimental model of SAH and administration of the specific CD34 antibody could prevent or reduce cerebral vasospasm caused by SAH.


Assuntos
Antígenos CD34/fisiologia , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD34/imunologia , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Vasoespasmo Intracraniano/etiologia
17.
Cell Signal ; 74: 109711, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32702440

RESUMO

The CD34 protein is regarded as a marker of stem cells from multiple origins. Recently a mesenchymal progenitor CD34 positive cell identified from traumatized human skeletal muscle demonstrates differentiation capability into vascular endothelial cells, osteoblasts and adipocytes. Here they were treated with a small inhibitory RNA for CD34, which significantly reduced the cellular level of the CD34 protein. These treated cells had a reduced capacity to proliferate, and migrate. They were both unable to differentiation down multiple pathways and to undergo vascular endothelial differentiation as reflected by a lack of expression of VE cadherin, Tie 2 and CD31. Additionally the cells were unable to form tube-like structures in an endothelial tube assay. These treated cells were also unable to undergo osteogenesis, as revealed by lack of alizarin red and alkaline phosphatase staining and were unable to undergo adipogenesis as revealed by lack of oil red O staining. Finally, when CD34 was expressed in cells lacking this protein, the cells were able to undergo vascular endothelial differentiation as revealed by expression of Tie2, VE-cadherin and CD31. These data indicate that in cells derived from traumatized muscle the CD34 protein is required for enhanced proliferation, migration and differentiation down multiple pathways.


Assuntos
Antígenos CD34/fisiologia , Células-Tronco Mesenquimais , Músculo Esquelético , Adipócitos/citologia , Adipogenia , Diferenciação Celular , Movimento Celular , Células Cultivadas , Células Endoteliais/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células Musculares/citologia , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Osteoblastos/citologia , Osteogênese
18.
Microcirculation ; 16(6): 487-96, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19479621

RESUMO

CD34 is a cell-surface sialomucin widely used for hematopoietic stem cell purification and as a marker of most vascular endothelial cells, including those of capillaries in the majority of tissues. Surprisingly, despite extensive research, the function of this sialomucin has remained elusive, with proposed roles ranging from enhancing proliferation or inhibiting differentiation to acting as a proadhesive L-selectin ligand. Here, we review our recent studies, which suggest that CD34 does, indeed, play a role in leukocyte and HSC trafficking, but that this is through its action as a regulated blocker of cell adhesion and enhancer of migration.


Assuntos
Antígenos CD34/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Antígenos CD34/química , Antígenos CD34/genética , Asma/etiologia , Biomarcadores , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/fisiologia , Adesão Celular/imunologia , Adesão Celular/fisiologia , Diferenciação Celular/imunologia , Diferenciação Celular/fisiologia , Movimento Celular/imunologia , Movimento Celular/fisiologia , Neoplasias do Colo/etiologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Mastócitos/citologia , Mastócitos/imunologia , Mastócitos/fisiologia , Camundongos , Microcirculação/imunologia , Microcirculação/fisiologia , Modelos Biológicos
19.
Stem Cells ; 26(4): 950-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18192237

RESUMO

CD34 is a transmembrane protein that is strongly expressed on hematopoietic stem/progenitor cells (HSCs); despite its importance as a marker of HSCs, its function is still poorly understood, although a role in cell adhesion has been demonstrated. To characterize the function of CD34 antigen on human HSCs, we examined, by both inhibition and overexpression, the role of CD34 in the regulation of HSC lineage differentiation. Our results demonstrate that CD34 silencing enhances HSC granulocyte and megakaryocyte differentiation and reduces erythroid maturation. In agreement with these results, the gene expression profile of these cells reveals the upregulation of genes involved in granulocyte and megakaryocyte differentiation and the downregulation of erythroid genes. Consistently, retroviral-mediated CD34 overexpression leads to a remarkable increase in erythroid progenitors and a dramatic decrease in granulocyte progenitors, as evaluated by clonogenic assay. Together, these data indicate that the CD34 molecule promotes the differentiation of CD34+ hematopoietic progenitors toward the erythroid lineage, which is achieved, at least in part, at the expense of granulocyte and megakaryocyte lineages.


Assuntos
Antígenos CD34/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Antígenos CD34/genética , Células Cultivadas , Células Clonais , Perfilação da Expressão Gênica , Inativação Gênica/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos
20.
Stem Cells ; 26(6): 1556-64, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18388301

RESUMO

Adipocytes are part of hematopoietic microenvironment, even though up to now in humans, their role in hematopoiesis is still questioned. We have previously shown that accumulation of fat cells in femoral bone marrow (BM) coincides with increased expression of neuropilin-1 (NP-1), while it is weakly expressed in hematopoietic iliac crest BM. Starting from this observation, we postulated that adipocytes might exert a negative effect on hematopoiesis mediated through NP-1. To test this hypothesis, we set up BM adipocytes differentiated into fibroblast-like fat cells (FLFC), which share the major characteristics of primitive unilocular fat cells, as an experimental model. As expected, FLFCs constitutively produced macrophage colony stimulating factor and induced CD34(+) differentiation into macrophages independently of cell-to-cell contact. By contrast, granulopoiesis was hampered by cell-to-cell contact but could be restored in transwell culture conditions, together with granulocyte colony stimulating factor production. Both functions were also recovered when FLFCs cultured in contact with CD34(+) cells were treated with an antibody neutralizing NP-1, which proved its critical implication in contact inhibition. An inflammatory cytokine such as interleukin-1 beta or dexamethasone modulates FLFC properties to restore granulopoiesis. Our data provide the first evidence that primary adipocytes exert regulatory functions during hematopoiesis that might be implicated in some pathological processes. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Adipócitos/fisiologia , Células da Medula Óssea/citologia , Fator Estimulador de Colônias de Granulócitos/antagonistas & inibidores , Neuropilina-1/fisiologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Animais , Antígenos CD/fisiologia , Antígenos CD34/fisiologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Primers do DNA , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Macrófagos/citologia , Macrófagos/fisiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA