Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
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
2.
Life Sci ; 231: 116521, 2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31152814

RESUMO

INTRODUCTION: Telocytes (TCs) are recently described to integrate a variety of different cells. AIM OF THE WORK: The aim was to investigate the presence of TCs in the rat mammary gland at its different physiological stages. MATERIAL AND METHODS: Twenty four adult female albino rats were classified into 4 groups: resting, mid-pregnancy, lactating, and involution groups. Inguinal mammary glands were processed for immunohistochemical and transmission electron microscopic (TEM) examination. RESULTS: TCs were immune-positive for c-kit and CD34 and showed significant differences in the different studied groups indicating variable roles at the different stages. TEM results characterized TCs by its shape and the long slender and moniliform telopodes linking the cells into stromal networks. The extracellular exosomes, homo-cellular synapsis and hetero-cellular synapsis were observed. CONCLUSION: Our study provides evidence for the presence of TCs in all stages of the gland; not only in the resting stage as proved by other studies, but with immune-labeling differences suggesting different structural and physiological roles of TCs according to the stage requirements. These functions might via controlling the proliferation during pregnancy and lactation and the involution of the gland after weaning. Thus, more future functional studies of TCs will be important to help understanding the mechanism by which TCs contribute to tissue homeostasis concerning the role of the stromal/epithelial interactions in mammary gland biology and pathology including breast cancer which would be revolutionary for future therapeutic applications.


Assuntos
Glândulas Mamárias Animais/fisiologia , Telócitos/fisiologia , Telócitos/ultraestrutura , Animais , Antígenos CD34/metabolismo , Antígenos CD34/fisiologia , Tecido Conjuntivo , Feminino , Imuno-Histoquímica , Lactação , Glândulas Mamárias Animais/citologia , Microscopia Eletrônica de Transmissão , Gravidez , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas c-kit/fisiologia , Ratos , Ratos Wistar
3.
Aging (Albany NY) ; 10(9): 2459-2479, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30243022

RESUMO

BACKGROUND: Heart failure (HF) is a major public health problem worldwide. The development of HF was related to coronary microvessel dysfunction. Whether miRNAs participate in HF by regulating coronary microvessel function remain unclear. METHODS: The potential targets of miR-665 were predicted by rnahybrid software, then verified through anti-Ago2 co-immunoprecipitation, Western blotting and luciferase reporter assays. rAAV9 system was used to manipulate the expression of miR-665 in vivo. RESULTS: Significant increase of miR-665 was observed in endothelial cells of human heart with heart failure. In vitro over-expression of miR-665 in endothelial cells resulted in decreased proliferation but enhanced apoptosis. rAAV-mediated delivery of miR-665 reduced coronary microvessel angiogenesis and cardiac microvessel density, then further impaired cardiac function in vivo. Furthermore, CD34 was confirmed as one of the miR-665 targets. Consistently, re-expression of CD34 attenuated miR-665-mediated damage effects in vitro and in vivo. We also found that Sp1 regulated miR-665 expression in endothelial cells. CONCLUSION: Our findings demonstrated that miR-665 played an important role in heart failure via damaging coronary microvessel angiogenesis, and suggested that miRNA-based therapeutics may protect against coronary microvessel dysfunction and heart failure.


Assuntos
Antígenos CD34/fisiologia , Vasos Coronários/fisiologia , Insuficiência Cardíaca/etiologia , MicroRNAs/fisiologia , Neovascularização Fisiológica/fisiologia , Animais , Endotélio Vascular/fisiologia , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/fisiologia , Fator de Transcrição Sp1/fisiologia
4.
PLoS One ; 12(8): e0182532, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28792982

RESUMO

The recruitment and retention of Natural Killer (NK) cells in the liver are thought to play an important role during hepatotropic infections and liver cirrhosis. The aims of this study were to determine differences between liver-derived and peripheral blood-derived NK cells in the context of liver inflammation and cirrhosis. We conducted a prospective dual-center cross-sectional study in patients undergoing liver transplantation or tumor-free liver resections, in which both liver tissue and peripheral blood samples were obtained from each consenting study participants. Intrahepatic lymphocytes and PBMCs were stained, fixed and analyzed by flow cytometry. Our results showed that, within cirrhotic liver samples, intrahepatic NK cells were particularly enriched for CD49a+ NK cells when compared to tumor-free liver resection samples. CD49a+ liver-derived NK cells included populations of cells expressing CD25, CD34 and CXCR3. Moreover, CD49a+CD25+ liver-derived NK cells exhibited high proliferative capacity in vitro in response to low doses of IL-2. Our study identified a specific subset of CD49a+CD25+ NK cells in cirrhotic livers bearing functional features of proliferation.


Assuntos
Proliferação de Células/fisiologia , Integrina alfa1/fisiologia , Subunidade alfa de Receptor de Interleucina-2/fisiologia , Células Matadoras Naturais/fisiologia , Fígado/citologia , Adulto , Idoso , Antígenos CD34/fisiologia , Estudos Transversais , Feminino , Citometria de Fluxo , Humanos , Células Matadoras Naturais/imunologia , Fígado/imunologia , Fígado/fisiologia , Transplante de Fígado , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Receptores CXCR3/fisiologia
5.
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
6.
Acta Cir Bras ; 31(1): 59-66, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26840357

RESUMO

PURPOSE: To describe a new technique for isolation of a mesenchymal stem cells (MSCs) population from the olfactory mucosa in rabbits. METHODS: Olfactory stem cells (OSCs) were retrieved from under the cribriform plate of the Ethmoid bone. Several assays were accomplished to characterize the cell population and attest its viability in vitro. The cells were submitted to flow cytometry with the antibodies CD34, CD45, CD73, CD79, CD90 and CD105 and also they were induced to differentiate in three lineages. Functional evaluation involved analysis of in vitro growth behavior, colony forming unit like fibroblasts (CFU-f) and cryopreservation response. Further transduction with Green Fluorescent Protein (GFP) was also performed. RESULTS: The OSCs showed mesenchymal features, as positive response to CD34, CD73 and CD90 antibodies and plasticity. Additionally, these cells have high proliferated rate, and they could be cultured through many passages and kept the ability to proliferate and differentiate after cryopreservation. The positive response to the transduction signalizes the possibility of cellular tracking in vivo. This is a desirable feature in case those cells are used for pre-clinical trials. CONCLUSION: The cells harvested were mesenchymal stem cells and the technique described is therefore efficient for rabbit olfactory stem cells isolation.


Assuntos
Separação Celular/métodos , Células-Tronco Mesenquimais/citologia , Mucosa Olfatória/citologia , 5'-Nucleotidase/fisiologia , Animais , Antígenos CD34/fisiologia , Diferenciação Celular/fisiologia , Plasticidade Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Criopreservação , Osso Etmoide/citologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Mucosa Olfatória/crescimento & desenvolvimento , Coelhos , Antígenos Thy-1/fisiologia
7.
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
8.
Stem Cell Res ; 15(3): 449-458, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26413784

RESUMO

Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) and their progenitors have been identified based on retrospective functional criteria. CD markers are employed to define cell populations with distinct functional characteristics. However, defining and prospective isolation of BMSCs and committed progenitors are lacking. Here, we compared the transcriptome profile of CD markers expressed at baseline and during the course of osteoblast and adipocyte differentiation of two well-characterized osteogenic-committed murine BMSCs (mBMSC(Bone)) and adipogenic-committed mBMSCs (mBMSC(Adipo)), respectively. Bioinformatic analysis revealed the presence of a core set of canonical mBMSC CD markers with comparable expression levels in mBMSC(Bone) and mBMSC(Adipo) at baseline and during their differentiation. We identified 11 CD markers that are differentially expressed between mBMSC(Adipo) and mBMSC(Bone). Among these, we identified osteoprogenitor-associated CD markers expressed only in mBMSC(Bone): CD34, CD54, CD73, CD132, CD200, CD227 and adipoprogenitor-associated CD markers expressed only in mBMSC(Adipo): CD53, CD80, CD134, CD141 and CD212. FACS analysis confirmed these results. We selected CD34 for further analysis. CD34 was expressed at baseline of mouse stromal cell line ST2, primary mBMSCs, mBMSC(Bone) and its expression decreased during osteoblast differentiation. FACS-sorted CD34(+) primary mBMSCs exhibited higher expression of 70% osteoblast-associated genes, and formed significantly higher heterotopic bone in vivo when implanted subcutaneously in immune-deficient mice compared with CD34(-) primary mBMSCs. Our results demonstrate that a set of CD markers can distinguish osteoprogenitor versus adipoprogenitor populations of mBMSCs. CD34 is suitable for prospective isolation of mouse bone marrow osteoprogenitors.


Assuntos
Antígenos CD34/fisiologia , Células da Medula Óssea/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Camundongos , Osteoblastos/citologia , Osteogênese/efeitos dos fármacos
9.
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
10.
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
11.
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
12.
Hypertens Res ; 34(9): 1017-22, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21654754

RESUMO

Angiotensin II (Ang II) is essential for endothelial progenitor cells (EPCs) function as Ang-II-induced oxidative stress causes senescence of EPCs and endothelial dysfunction and Ang II type 1 receptor blockers increase EPCs. Moreover, EPCs activity is dependent on nitric oxide (NO) and heme oxygenase (HO)-1 as these correlate with EPCs senescence and are reduced in hypertensives. Bartter's/Gitelman's syndrome patients (BS/GS), have increased Ang II yet normo/hypotension along with blunted Ang II signaling, reduced oxidative stress, increased NO and HO-1, thus presenting a unique system to explore EPC biology and its relationship with vascular clinical and biochemical correlates. Circulating EPCs, NO-dependent vasodilation (flow-mediated dilation (FMD)) and HO-1 gene expression were characterized in 10 BS/GS patients and in 10 normotensive subjects. EPCs defined by cell surface antigens CD34+kinase-insert domain receptor (KDR+), CD133+KDR+ and CD133+CD34+KDR+ cells were quantitiated via direct three-color flow-cytometry analysis, HO-1 gene expression by reverse transcription-PCR and FMD by B-mode echo scan of the right brachial artery. Correlation analysis was carried out regarding FMD and EPCs, FMD and HO-1 and EPCs and HO-1. In BS/GS, CD34+KDR+ cell numbers did not differ from controls while CD133+KDR+ and CD133+CD34+KDR+ cell numbers were higher. HO-1 gene expression, as well as FMD, was higher in BS/GS compared with controls. Both CD133+KDR+ and CD133+CD34+KDR+ strongly correlated with both FMD and HO-1. FMD and HO-1 were also strongly correlated. These results document in a human system that EPC numbers and specific populations are related to important clinical and biochemical factors involved in cardiovascular (CV) status and reaffirm the utility of BS/GS patients as a useful system to investigate EPC's role(s) in the pathophysiology of cardiovascular remodeling in humans.


Assuntos
Angiotensina II/fisiologia , Síndrome de Bartter/fisiopatologia , Células Endoteliais/fisiologia , Síndrome de Gitelman/fisiopatologia , Células-Tronco/fisiologia , Antígeno AC133 , Adulto , Antígenos CD/fisiologia , Antígenos CD34/fisiologia , Síndrome de Bartter/genética , Artéria Braquial/fisiologia , Artéria Braquial/fisiopatologia , Feminino , Síndrome de Gitelman/genética , Glicoproteínas/fisiologia , Heme Oxigenase-1/genética , Heme Oxigenase-1/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/fisiologia , Peptídeos/fisiologia , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Vasodilatação/fisiologia
13.
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
14.
J Gerontol A Biol Sci Med Sci ; 65(10): 1042-50, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20591876

RESUMO

Changes in progenitor cell biology remain at the forefront of many theories of biologic aging, but there are limited studies evaluating this in humans. Aging has been associated with a progressive depletion of circulating progenitor cells, but age-related bone marrow-resident progenitor cell depletion has not been systematically determined in humans. Patients undergoing total hip replacement were consented, and bone marrow and peripheral progenitor cells were enumerated based on aldehyde dehydrogenase activity and CD34 and CD133 expression. Circulating progenitors demonstrated an age-dependent decline. In contrast, marrow-resident progenitor cell content demonstrated no age association with any progenitor cell subtype. In humans, aging is associated with depletion of circulating, but not marrow-resident, progenitors. This finding has impact on the mechanism(s) responsible for age-related changes in circulating stem cells and important implications for the use of autologous marrow for the treatment of age-related diseases.


Assuntos
Envelhecimento/fisiologia , Células da Medula Óssea/fisiologia , Células-Tronco/fisiologia , Antígeno AC133 , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Aldeído Desidrogenase/fisiologia , Antígenos CD/fisiologia , Antígenos CD34/fisiologia , Antígenos de Superfície/fisiologia , Contagem de Células Sanguíneas , Feminino , Citometria de Fluxo , Glicoproteínas/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeos/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Adulto Jovem
15.
Diabetes ; 59(8): 2010-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20460428

RESUMO

OBJECTIVE: Peripheral blood CD34(+) cells from diabetic patients demonstrate reduced vascular reparative function due to decreased proliferation and diminished migratory prowess, largely resulting from decreased nitric oxide (NO) bioavailability. The level of TGF-beta, a key factor that modulates stem cell quiescence, is increased in the serum of type 2 diabetic patients. We asked whether transient TGF-beta1 inhibition in CD34(+) cells would improve their reparative ability. RESEARCH DESIGN AND METHODS: To inhibit TGF-beta1 protein expression, CD34(+) cells were treated ex vivo with antisense phosphorodiamidate morpholino oligomers (TGF-beta1-PMOs) and analyzed for cell surface CXCR4 expression, cell survival in the absence of added growth factors, SDF-1-induced migration, NO release, and in vivo retinal vascular reparative ability. RESULTS: TGF-beta1-PMO treatment of diabetic CD34(+) cells resulted in increased expression of CXCR4, enhanced survival in the absence of growth factors, and increased migration and NO release as compared with cells treated with control PMO. Using a retinal ischemia reperfusion injury model in mice, we observed that recruitment of diabetic CD34(+) cells to injured acellular retinal capillaries was greater after TGF-beta1-PMO treatment compared with control PMO-treated cells. CONCLUSIONS: Transient inhibition of TGF-beta1 may represent a promising therapeutic strategy for restoring the reparative capacity of dysfunctional diabetic CD34(+) cells.


Assuntos
Diabetes Mellitus/fisiopatologia , Angiopatias Diabéticas/prevenção & controle , Células-Tronco Hematopoéticas/fisiologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos CD34/metabolismo , Antígenos CD34/fisiologia , Capilares/fisiopatologia , Sobrevivência Celular , Retinopatia Diabética/fisiopatologia , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Morfolinos , Óxido Nítrico/metabolismo , Receptores CXCR4/genética , Traumatismo por Reperfusão/fisiopatologia , Fator de Crescimento Transformador beta1/metabolismo
16.
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
17.
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
18.
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
19.
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
20.
Transplant Proc ; 41(5): 1885-90, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19545750

RESUMO

BACKGROUND: There is an increasing need for suitable animal models for the study of the human immune system and disease. The purpose of this study was to develop a practical in vivo model of human immune cell repopulation using ex vivo expanded human fetal liver-derived CD34(+) hematopoietic stem cells and subrenally coimplanted fetal liver/thymus tissues. METHODS: Freshly isolated fetal liver-derived CD34(+) hematopoietic stem cells were frozen until injected and ex vivo expanded with various cytokines for 7 days. After fetal liver/thymus tissues were subrenally coimplanted into preirradiated Rag2(-/-)gamma(c)(-/-) mice, frozen and ex vivo expanded CD34(+) cells were injected intravenously. The peripheral blood of the mice was monitored for the detection of human cell engraftment using flow cytometry. Then we confirmed human T-cell function by in vitro function assays. RESULTS: After fetal liver/thymus tissues were coimplanted into the irradiated Rag2(-/-)gamma(c)(-/-) mice, with frozen and ex vivo expanded CD34(+) hematopoietic stem cells, human cell engraftments were determined using hCD45 and multilineage markers. The cultured cells with the cytokine combination of stem cell factor, thrombopoietin, Flk2/Flk3 ligand (FL), and interleukin-3 showed stable and long-term engraftment compared to other combinations. The ex vivo expanded human fetal liver-derived CD34(+) hematopoietic stem cells, under our culture conditions, accomplished a large volume of expanded cells that were sustained, demonstrating self-renewal of the evaluated markers, which may have indicated long- term repopulation activity. CONCLUSION: The results of this study demonstrated a practical mouse model of expanded human immune cells especially T cells in Rag2(-/-)gamma(c)(-/-) mice.


Assuntos
Transplante de Fígado/imunologia , Timo/transplante , Animais , Antígenos CD34/fisiologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Transplante de Tecido Fetal , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Fígado/embriologia , Transfusão de Linfócitos , Camundongos , Camundongos Knockout , Camundongos SCID , Ensaio de Cápsula Sub-Renal , Timo/embriologia , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA