Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
Mediators Inflamm ; 2018: 2691934, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30116144

RESUMO

BACKGROUND: Acute myocardial infarction (AMI) and coronary artery bypass graft (CABG) surgery are associated with a pathogen-free inflammatory response (sterile inflammation). Complement cascade (CC) and bioactive sphingolipids (BS) are postulated to be involved in this process. AIM: The aim of this study was to evaluate plasma levels of CC cleavage fragments (C3a, C5a, and C5b9), sphingosine (SP), sphingosine-1-phosphate (S1P), and free hemoglobin (fHb) in AMI patients treated with primary percutaneous coronary intervention (pPCI) and stable coronary artery disease (SCAD) undergoing CABG. PATIENTS AND METHODS: The study enrolled 37 subjects (27 male) including 22 AMI patients, 7 CABG patients, and 8 healthy individuals as the control group (CTRL). In the AMI group, blood samples were collected at 5 time points (admission to hospital, 6, 12, 24, and 48 hours post pPCI) and 4 time points in the CABG group (6, 12, 24, and 48 hours post operation). SP and S1P concentrations were measured by high-performance liquid chromatography (HPLC). Analysis of C3a, C5a, and C5b9 levels was carried out using high-sensitivity ELISA and free hemoglobin by spectrophotometry. RESULTS: The plasma levels of CC cleavage fragments (C3a and C5b9) were significantly higher, while those of SP and S1P were lower in patients undergoing CABG surgery in comparison to the AMI group. In both groups, levels of CC factors showed no significant changes within 48 hours of follow-up. Conversely, SP and S1P levels gradually decreased throughout 48 hours in the AMI group but remained stable after CABG. Moreover, the fHb concentration was significantly higher after 24 and 48 hours post pPCI compared to the corresponding postoperative time points. Additionally, the fHb concentrations increased between 12 and 48 hours after PCI in patients with AMI. CONCLUSIONS: Inflammatory response after AMI and CABG differed regarding the release of sphingolipids, free hemoglobin, and complement cascade cleavage fragments.


Assuntos
Proteínas do Sistema Complemento/análise , Doença da Artéria Coronariana/sangue , Hemoglobinas/análise , Infarto do Miocárdio/sangue , Esfingolipídeos/metabolismo , Idoso , Estudos de Casos e Controles , Ponte de Artéria Coronária , Feminino , Humanos , Inflamação , Lisofosfolipídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Intervenção Coronária Percutânea , Esfingolipídeos/sangue , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Resultado do Tratamento
2.
Am J Transplant ; 16(2): 440-53, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26550777

RESUMO

CD8(+)/TCR(-) facilitating cells (FCs) in mouse bone marrow (BM) significantly enhance engraftment of hematopoietic stem/progenitor cells (HSPCs). Human FC phenotype and mechanism of action remain to be defined. We report, for the first time, the phenotypic characterization of human FCs and correlation of phenotype with function. Approximately half of human FCs are CD8(+)/TCR(-)/CD56 negative (CD56(neg)); the remainder are CD8(+)/TCR(-)/CD56 bright (CD56(bright)). The CD56(neg) FC subpopulation significantly promotes homing of HSPCs to BM in nonobese diabetic/severe combined immunodeficiency/IL-2 receptor γ-chain knockout mouse recipients and enhances hematopoietic colony formation in vitro. The CD56(neg) FC subpopulation promotes rapid reconstitution of donor HSPCs without graft-versus-host disease (GVHD); recipients of CD56(bright) FCs plus HSPCs exhibit low donor chimerism early after transplantation, but the level of chimerism significantly increases with time. Recipients of HSPCs plus CD56(neg) or CD56(bright) FCs showed durable donor chimerism at significantly higher levels in BM. The majority of both FC subpopulations express CXCR4. Coculture of CD56(bright) FCs with HSPCs upregulates cathelicidin and ß-defensin 2, factors that prime responsiveness of HSPCs to stromal cell-derived factor 1. Both FC subpopulations significantly upregulated mRNA expression of the HSPC growth factors and Flt3 ligand. These results indicate that human FCs exert a direct effect on HSPCs to enhance engraftment. Human FCs offer a potential regulatory cell-based therapy for enhancement of engraftment and prevention of GVHD.


Assuntos
Antígenos CD8/metabolismo , Doença Enxerto-Hospedeiro/imunologia , Células-Tronco Hematopoéticas/imunologia , Subunidade gama Comum de Receptores de Interleucina/fisiologia , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Apoptose , Western Blotting , Células Cultivadas , Doença Enxerto-Hospedeiro/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Modelos Animais , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Doadores de Tecidos , Quimeras de Transplante
3.
J Exp Med ; 175(3): 743-50, 1992 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-1371302

RESUMO

To determine whether N-ras expression is required at specific stages of the process of in vitro normal human hematopoiesis, adherent- and T lymphocyte-depleted mononuclear marrow cells (A-T-MNC) or highly purified progenitors (CD34+ cells) were cultured in semisolid medium, under conditions that favor the growth of specific progenitor cell types, after exposure to N-ras sense and antisense oligodeoxynucleotides. N-ras antisense, but not sense, oligodeoxynucleotide treatment of A-T-MNC and CD34+ cells resulted in a significantly decreased number of granulocyte/macrophage colony-forming units (CFU-GM) induced by interleukin 3 (IL-3) or granulocyte/macrophage colony-stimulating factor (GM-CSF) and of macrophage colonies (CFU-M) induced by M-CSF, but not of granulocytic colonies induced with G-CSF or IL-5. However, the same treatment significantly inhibited colony formation induced by each of the above factors in combination with IL-3. Megakaryocytic colony (CFU-Meg) formation from A-T-MNC or CD34+ cells in the presence of IL-6 + IL-3 + erythropoietin (Epo) was also markedly decreased after antisense oligodeoxynucleotide treatment. Erythroid colonies derived from A-T-MNC in the presence of Epo (CFU-E) were not inhibited upon antisense treatment, whereas those arising from A-T-MNC or CD34+ cells in the presence of IL-3 + Epo (BFU-E) were markedly affected. These results are consistent with the hypothesis that distinct signal transduction pathways, involving N-ras or not, are activated by different growth factors in different hematopoietic progenitor cells.


Assuntos
Substâncias de Crescimento/farmacologia , Hematopoese/efeitos dos fármacos , Oligonucleotídeos Antissenso/genética , Antígenos CD/análise , Antígenos CD34 , Sequência de Bases , Medula Óssea/imunologia , Células da Medula Óssea , Depressão Química , Células Precursoras Eritroides/efeitos dos fármacos , Genes ras/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos dos fármacos , Humanos , Megacariócitos/citologia , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/farmacologia , RNA Mensageiro/análise , Células-Tronco/efeitos dos fármacos , Transcrição Gênica
4.
Herz ; 35(7): 467-72, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20981396

RESUMO

The bone marrow (BM) niche contains small heterogenous populations of cells which may contribute to cardiac and endothelial repair, including committed lineages [endothelial progenitor cells (EPCs), multipotent mesenchymal stromal cells (MSCs) and more primitive very small embryonic-like cells (VSELs) expressing pluripotent stem cell (PSC) markers (Oct-4, Nanog, SSEA-1)]. VSELs are present in BM, peripheral blood and some solid organs in mice and were recently identified in peripheral blood in patients with acute coronary syndromes and stroke. VSELs can be expanded in vitro and differentiated into cells from all three germ layers. This population of cells displays the morphology of primitive PSC (small size, open type chromatin, large nucleus, narrow rim of cytoplasm) and express PSC markers. The isolation of human VSELs is based on their size and presence of several surface markers (CXCR4, CD133, CD34) and lack of markers of hematopoietic lineage (lin, CD45). In acute myocardial infarction and ischemic stroke VSELs are rapidly mobilized into peripheral blood, and express increased levels of PSC markers as well as early cardiac (GATA-4, Nkx2.5/Csx), neural (GFAP, nestin, beta-III-tubulin, Olig1, Olig2, Sox2, Musashi) and endothelial lineage markers (VE-cadherin, von Willebrand factor). The number of VSELs mobilized in acute myocardial infarction is inversely correlated with left ventricular ejection fraction and the release of cardiac necrosis markers. Mobilization of these cells is also reduced in patients with diabetes and in the elderly. BM-derived VSELs were expanded and after cardiogenic pre-differentiation injected intramyocardially in mice models of myocardial infarction leading to improved left ventricular contractility. VSELs are probably progeny of epiblast cells which migrated to the BM and developing organs during embryonic development. The cells are present in a quiescent state in the adult BM and solid organs and might serve as a reserve pool of resident stem cells. VSELs are promising candidates for further pre-clinical and clinical studies on cellular cardiovascular therapy.


Assuntos
Síndrome Coronariana Aguda/cirurgia , Células-Tronco Embrionárias/transplante , Miócitos Cardíacos/transplante , Acidente Vascular Cerebral/cirurgia , Engenharia Tecidual/métodos , Síndrome Coronariana Aguda/complicações , Síndrome Coronariana Aguda/patologia , Animais , Células-Tronco Embrionárias/patologia , Humanos , Camundongos , Miócitos Cardíacos/patologia , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia
5.
Stem Cell Rev Rep ; 16(1): 198-211, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31828580

RESUMO

BACKGROUND: Bone marrow-derived mononuclear cells (BM-MNC) consist of a heterogeneous mix of mesenchymal stem cells (MSC), hematopoietic progenitor cells (HPC), endothelial progenitor cells (EPC), monocytes, lymphocytes and pluripotent stem cells. Whereas the importance of MSC and EPC has been well documented in bone healing and regeneration studies, the role of pluripotent stem cells is still poorly understood. In the present study we evaluated if and how Very Small Embryonic Like cells (VSEL), isolated from rat BM-MNC, contribute to bone healing. METHODS: Large bone defects were made in the femurs of 38 Sprague Dawley female rats and treated with ß-TCP scaffold granules seeded with male VSEL; BM-MNC, VSEL-depleted BM-MNC or scaffold alone, and bone healing was evaluated at 8 weeks post-surgery. RESULTS: Bone healing was significantly increased in defects treated with VSEL and BM-MNC, compared to defects treated with VSEL-depleted BM-MNC. Donor cells were detected in new bone tissue, in all the defects treated with cells, and in fibrous tissue only in defects treated with VSEL-depleted BM-MNC. The number of CD68+ cells was the highest in the VSEL-depleted group, whereas the number of TRAP positive cells was the lowest in this group. CONCLUSIONS: Based on the results, we can conclude that VSEL play a role in BM-MNC induced bone formation. In our rat femur defect model, in defects treated with VSEL-depleted BM-MNC, osteoclastogenesis and bone formation were decreased, and foreign body reaction was increased.


Assuntos
Células-Tronco Adultas/transplante , Regeneração Óssea/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Pluripotentes/transplante , Adulto , Animais , Células Progenitoras Endoteliais/transplante , Humanos , Monócitos/transplante , Osteogênese/genética , Ratos
6.
Mech Ageing Dev ; 130(1-2): 58-66, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18377952

RESUMO

Recently our group identified in murine bone marrow (BM) and human cord blood (CB), a rare population of very small embryonic-like (VSEL) stem cells. We hypothesize that these cells are deposited during embryonic development in BM as a mobile pool of circulating pluripotent stem cells (PSC) that play a pivotal role in postnatal tissue turnover both of non-hematopoietic and hematopoietic tissues. During in vitro co-cultures with murine myoblastic C2C12 cells, VSELs form spheres that contain primitive stem cells. Cells isolated from these spheres may give rise to cells from all three germ layers when plated in tissue specific media. The number of murine VSELs and their ability to form spheres decreases with the age and is reduced in short-living murine strains. Thus, developmental deposition of VSELs in adult tissues may potentially play an underappreciated role in regulating the rejuvenation of senescent organs. We envision that the regenerative potential of these cells could be harnessed to decelerate aging processes.


Assuntos
Envelhecimento/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Adulto , Idoso , Animais , Tamanho Celular , Senescência Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Humanos
7.
Cell Tissue Res ; 331(1): 125-34, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17828555

RESUMO

Bone marrow (BM) develops in mammals by the end of the second/beginning of the third trimester of gestation and becomes a major hematopoietic organ in postnatal life. The alpha-chemokine stromal derived factor-1 (SDF-1) to CXCR4 (G ai-protein-coupled seven transmembrane-spanning chemokine receptor) axis plays a major role in BM colonization by stem cells. By the end of the second trimester of gestation, BM becomes colonized by hematopoietic stem cells (HSC), which are chemoattracted from the fetal liver in a CXCR4-SDF-1-dependent manner. Whereas CXCR4 is expressed on HSC, SDF-1 is secreted by BM stroma and osteoblasts that line BM cavities. Mounting evidence indicates that BM also contains rare CXCR4(+) pluripotent stem cells (PSC). Recently, our group has identified a population of CXCR4(+) very small embryonic like stem cells in murine BM and human cord blood. We hypothesize that these cells are deposited during development in BM as a mobile pool of circulating PSC that play a pivotal role in postnatal tissue turnover, both of non-hematopoietic and hematopoietic tissues.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Embrionárias/citologia , Animais , Impressão Genômica , Células Germinativas , Humanos , Células-Tronco Pluripotentes/citologia
8.
Cytotherapy ; 10(2): 182-92, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18368597

RESUMO

BACKGROUND: In patients transplanted with cord blood (CB), prolonged thrombocytopenia is a major complication. However, this could be alleviated by supplementing the CB graft with ex vivo-expanded megakaryocytic progenitors (CFU-Meg), provided that the homing properties of these cells are not affected negatively by expansion. METHODS AND RESULTS: We assessed the in vitro homing potential of CFU-Meg progenitors expanded from CB and showed that the combination of thrombopoietin (TPO) with interleukin-3 (IL-3) used for expansion not only results in optimal proliferation of CFU-Meg but also protects these cells from apoptosis. Moreover, we found that ex vivo-expanded CFU-Meg maintained expression of the CXCR4 receptor throughout a 9-day culture and were chemoattracted towards a stromal cell-derived factor-1 (SDF-1) gradient. They also expressed matrix metalloproteinase-9 (MMP-9) and membrane-type (MT) 1-MMP, and transmigrated across the reconstituted basement membrane Matrigel. Finally, we observed that SDF-1 up-regulated the expression of both MMP-9 and MT1-MMP in CB CD34(+) cells and ex vivo-expanded CFU-Meg. DISCUSSION: We suggest that CB-expanded CFU-Meg, in particular those from day 3 of expansion, when their proliferation and in vitro homing potential are maximal, could be employed to supplement CB grafts and speed up platelet recovery in transplant recipients.


Assuntos
Ensaio de Unidades Formadoras de Colônias , Sangue Fetal/citologia , Sangue Fetal/enzimologia , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Megacariócitos/citologia , Células-Tronco/citologia , Antígenos CD34/metabolismo , Catequina/análogos & derivados , Catequina/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Fatores Quimiotáticos/farmacologia , Quimiotaxia/efeitos dos fármacos , Colágeno/metabolismo , Combinação de Medicamentos , Sangue Fetal/efeitos dos fármacos , Humanos , Interleucina-3/farmacologia , Cinética , Laminina/metabolismo , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Megacariócitos/efeitos dos fármacos , Megacariócitos/enzimologia , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Proteoglicanas/metabolismo , Receptores CXCR4/metabolismo , Células-Tronco/efeitos dos fármacos , Trombopoetina/farmacologia , Regulação para Cima/efeitos dos fármacos
9.
Leukemia ; 21(5): 973-82, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17330096

RESUMO

We hypothesized that the third complement component (C3) cleavage fragments (C3a and (des-Arg)C3a) are involved in stress/inflammation-related thrombocytosis, and investigated their potential role in reactive thrombocytosis induced by bleeding. We found that platelet counts are lower in C3-deficient mice in response to excessive bleeding as compared to normal littermates and that C3a and (des-Arg)C3a enhance stromal-derived factor-1 (SDF-1)-dependent megakaryocyte (Megs) migration, adhesion and platelet shedding. At the molecular level, C3a stimulates in Megs MAPKp42/44 phosphorylation, and enhances incorporation of CXCR4 into membrane lipid rafts increasing the responsiveness of Megs to SDF-1. We found that perturbation of lipid raft formation by statins decreases SDF-1/C3a-dependent platelet production in vitro and in an in vivo model statins ameliorated post-bleeding thrombocytosis. Thus, inhibition of lipid raft formation could find potential clinical application as a means of ameliorating some forms of thrombocytosis.


Assuntos
Quimiocinas CXC/fisiologia , Complemento C3a/fisiologia , Hemorragia/fisiopatologia , Trombocitose/fisiopatologia , Trombopoese , Quimiocina CXCL12 , Humanos , Microdomínios da Membrana/fisiologia , Contagem de Plaquetas , Receptores CXCR4/fisiologia , Receptores de Complemento/análise , Transdução de Sinais
10.
Leukemia ; 21(5): 860-7, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17344915

RESUMO

Accumulating evidence demonstrates that adult tissues contain a population of stem cells that express early developmental markers such as stage-specific embryonic antigen and transcription factors Oct-4 and Nanog. These are the markers characteristic for embryonic stem cells, epiblast stem cells and primordial germ cells. The presence of these stem cells in adult tissues including bone marrow, epidermis, bronchial epithelium, myocardium, pancreas and testes supports the concept that adult tissues contain some population of pluripotent stem cells that is deposited in embryogenesis during early gastrulation. In this review we will discuss these data and present a hypothesis that these cells could be direct descendants of the germ lineage. The germ lineage in order to pass genes on to the next generations creates soma and thus becomes a 'mother lineage' for all somatic cell lineages present in the adult body.


Assuntos
Células-Tronco Adultas/química , Células da Medula Óssea/citologia , Células-Tronco Embrionárias/citologia , Fator 3 de Transcrição de Octâmero/análise , Células-Tronco Pluripotentes/citologia , Animais , Linhagem da Célula , Humanos
11.
Leukemia ; 21(2): 297-303, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17136117

RESUMO

Recently, we purified from adult murine bone marrow (BM) a population of CXCR4(+), Oct-4(+) SSEA-1(+), Sca-1(+) lin(-) CD45(-) very small embryonic-like (VSEL) stem cells and hypothesized that similar cells could be also present in human cord blood (CB). Here, we report that by employing a novel two-step isolation procedure -- removal of erythrocytes by hypotonic lysis combined with multiparameter sorting -- we could isolate from CB a population of human cells that are similar to murine BM-derived VSELs, described previously by us. These CB-isolated VSELs (CB-VSEL) are very small (3-5 micro m) and highly enriched in a population of CXCR4(+)AC133(+)CD34(+)lin(-) CD45(-) CB mononuclear cells, possess large nuclei containing unorganized euchromatin and express nuclear embryonic transcription factors Oct-4 and Nanog and surface embryonic antigen SSEA-4. Further studies are needed to see if human CB-isolated VSELs similar to their murine BM-derived counterparts are endowed with pluripotent stem cell properties.


Assuntos
Células-Tronco Embrionárias/citologia , Sangue Fetal/citologia , Glicoesfingolipídeos/genética , Fator 3 de Transcrição de Octâmero/genética , Receptores CXCR4/genética , Animais , Células da Medula Óssea/citologia , Tamanho Celular , Células-Tronco Embrionárias/ultraestrutura , Humanos , Recém-Nascido , Camundongos , Microscopia Eletrônica , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Antígenos Embrionários Estágio-Específicos
12.
J Clin Invest ; 104(12): 1739-49, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10606628

RESUMO

CD34(+) cells are nonpermissive to infection by HIV strains X4 and R5, despite the fact that many CD34(+) cells express high levels of the viral receptor protein CD4 and the coreceptor CXCR4 on their surface. In these cells, the co-receptor CCR5 protein, which, like CXCR4, is a chemokine receptor, is detected mainly intracellularly. We hypothesized that CD34(+) cells secrete CCR5-binding chemokines and that these factors interfere with HIV R5 interactions with these cells, possibly by binding CCR5 or by inducing its internalization. We found that human CD34(+) cells and CD34(+)KIT(+) cells, which are enriched in myeloid progenitor cells, expressed and secreted the CCR5 ligands RANTES, MIP-1alpha, and MIP-1beta and that IFN-gamma stimulated expression of these chemokines. In contrast, SDF-1, a CXCR4 ligand, was not detectable in the CD34(+)KIT(+) cells, even by RT-PCR. Conditioned media from CD34(+) cell culture significantly protected the T lymphocyte cell line PB-1 from infection by R5 but not X4 strains of HIV. Interestingly, the secretion of endogenous chemokines decreased with the maturation of CD34(+) cells, although ex vivo, expanded megakaryoblasts still secreted a significant amount of RANTES. Synthesis of CCR5-binding chemokines by human CD34(+) cells and megakaryoblasts therefore largely determines the susceptibility of these cells to infection by R5 HIV strains. We postulate that therapeutic agents that induce the endogenous synthesis of chemokines in human hematopoietic cells may protect these cells from HIV infection.


Assuntos
Antígenos CD34/análise , Células da Medula Óssea/fisiologia , Quimiocinas/fisiologia , HIV/fisiologia , Células-Tronco Hematopoéticas/virologia , Megacariócitos/fisiologia , Quimiocinas/análise , Humanos , Interferon gama/farmacologia , RNA Mensageiro/análise , Receptores CCR5/análise , Receptores CXCR4/análise
13.
J Clin Invest ; 94(1): 320-7, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8040273

RESUMO

IGF-I has been reported to increase hematopoietic progenitor cell cloning efficiency. To investigate this phenomenon, we studied the IGF-I responsiveness of human marrow cells expressing IGF-I receptor (IGF-IR), a direct strategy not used previously. IGF-IR+ and control CD34+ marrow cells were isolated using immunoaffinity methods. Then, the cells were cloned in methylcellulose containing variable amounts of serum- and lineage-appropriate growth factors supplemented with recombinant human IGF-I. In contrast to CD34+ cells, IGF-IR+ cells never gave rise to CFU-Blast, CFU-Mix, CFU-GM, BFU-E, or CFU-E. To substantiate the suggestion that CD34+ and IGF-IR+ cells were distinct populations, we used reverse transcription PCR to detect IGF-I, EpO, and KIT receptor mRNAs in these cells. The mRNA phenotype of CD34+ cells was EpO (+), KIT (+), and IGF-IR (-), while IGF-IR+ cells were IGF-IR (+), EpO (-), and KIT (-). These results suggested that IGF-IR is either not expressed or expressed at low levels on normal hematopoietic progenitor cells. Functional significance of the latter possibility was tested by exposing CD34+ cells to IGF-IR antisense oligodeoxynucleotides. Colony formation was unaffected by oligodeoxynucleotide disruption of IGF-IR, suggesting that, even if expressed at low level, the receptor's functional significance was doubtful. Nevertheless, when cultured in the presence of IGF-I, IGF-IR+ cells elaborated an activity with mild BFU-E stimulatory effects. Accordingly, if IGF-I plays a role in hematopoietic colony formation, it is probably and results from stimulation of IGF-IR-positive ancillary cells to secrete growth factors. Studies carried out with human leukemia cells yielded similar results.


Assuntos
Hematopoese/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Leucemia/patologia , Dados de Sequência Molecular , Receptor IGF Tipo 1/análise
14.
Leukemia ; 20(9): 1487-95, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16791265

RESUMO

Normal and malignant cells shed from their surface membranes as well as secrete from the endosomal membrane compartment circular membrane fragments called microvesicles (MV). MV that are released from viable cells are usually smaller in size compared to the apoptotic bodies derived from damaged cells and unlike them do not contain fragmented DNA. Growing experimental evidence indicates that MV are an underappreciated component of the cell environment and play an important pleiotropic role in many biological processes. Generally, MV are enriched in various bioactive molecules and may (i) directly stimulate cells as a kind of 'signaling complex', (ii) transfer membrane receptors, proteins, mRNA and organelles (e.g., mitochondria) between cells and finally (iii) deliver infectious agents into cells (e.g., human immuno deficiency virus, prions). In this review, we discuss the pleiotropic effects of MV that are important for communication between cells, as well as the role of MV in carcinogenesis, coagulation, immune responses and modulation of susceptibility/infectability of cells to retroviruses or prions.


Assuntos
Comunicação Celular , Membrana Celular/fisiologia , Organelas/fisiologia , Progressão da Doença , Humanos , Neoplasias/patologia
15.
Leukemia ; 20(5): 857-69, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16498386

RESUMO

By employing multiparameter sorting, we identified in murine bone marrow (BM) a homogenous population of rare (approximately 0.02% of BMMNC) Sca-1(+)lin(-)CD45- cells that express by RQ-PCR and immunohistochemistry markers of pluripotent stem cells (PSC) such as SSEA-1, Oct-4, Nanog and Rex-1. The direct electronmicroscopical analysis revealed that these cells are small (approximately 2-4 microm), posses large nuclei surrounded by a narrow rim of cytoplasm, and contain open-type chromatin (euchromatin) that is typical for embryonic stem cells. In vitro cultures these cells are able to differentiate into all three germ-layer lineages. The number of these cells is highest in BM from young (approximately 1-month-old) mice and decreases with age. It is also significantly diminished in short living DBA/2J mice as compared to long living B6 animals. These cells in vitro respond strongly to SDF-1, HGF/SF and LIF and express CXCR4, c-met and LIF-R, respectively, and since they adhere to fibroblasts they may be coisolated with BM adherent cells. We hypothesize that this population of Sca-1(+)lin(-)CD45- very small embryonic-like (VSEL) stem cells is deposited early during development in BM and could be a source of pluripotent stem cells for tissue/organ regeneration.


Assuntos
Células da Medula Óssea/citologia , Antígenos CD15/biossíntese , Fator 3 de Transcrição de Octâmero/biossíntese , Receptores CXCR4/biossíntese , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores Etários , Animais , Diferenciação Celular/fisiologia , Separação Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Feminino , Técnicas In Vitro , Antígenos CD15/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Fator 3 de Transcrição de Octâmero/genética , Pâncreas/citologia , Pâncreas/fisiologia , Proteínas Proto-Oncogênicas c-met/genética , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Células-Tronco/classificação
16.
Leukemia ; 20(11): 1915-24, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16900209

RESUMO

Proper response of normal stem cells (NSC) to motomorphogens and chemoattractants plays a pivotal role in organ development and renewal/regeneration of damaged tissues. Similar chemoattractants may also regulate metastasis of cancer stem cells (CSC). Growing experimental evidence indicates that both NSC and CSC express G-protein-coupled seven-transmembrane span receptor CXCR4 and respond to its specific ligand alpha-chemokine stromal derived factor-1 (SDF-1), which is expressed by stroma cells from different tissues. In addition, a population of very small embryonic-like (VSEL) stem cells that express CXCR4 and respond robustly to an SDF-1 gradient was recently identified in adult tissues. VSELs express several markers of embryonic and primordial germ cells. It is proposed that these cells are deposited early in the development as a dormant pool of embryonic/pluripotent NSC. Expression of both CXCR4 and SDF-1 is upregulated in response to tissue hypoxia and damage signal attracting circulating NSC and CSC. Thus, pharmacological modulation of the SDF-1-CXCR4 axis may lead to the development of new therapeutic strategies to enhance mobilization of CXCR4+ NSC and their homing to damaged organs as well as inhibition of the metastasis of CXCR4+ cancer cells.


Assuntos
Quimiocinas CXC/fisiologia , Neoplasias/fisiopatologia , Organogênese/fisiologia , Receptores CXCR4/fisiologia , Regeneração/fisiologia , Animais , Quimiocina CXCL12 , Humanos
17.
Leukemia ; 20(5): 847-56, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16453000

RESUMO

Membrane-derived vesicles (MV) are released from the surface of activated eucaryotic cells and exert pleiotropic effects on surrounding cells. Since the maintenance of pluripotency and undifferentiated propagation of embryonic stem (ES) cells in vitro requires tight cell to cell contacts and effective intercellular signaling, we hypothesize that MV derived from ES cells (ES-MV) express stem cell-specific molecules that may also support self-renewal and expansion of adult stem cells. To address this hypothesis, we employed expansion of hematopoietic progenitor cells (HPC) as a model. We found that ES-MV (10 microg/ml) isolated from murine ES cells (ES-D3) in serum-free cultures significantly (i) enhanced survival and improved expansion of murine HPC, (ii) upregulated the expression of early pluripotent (Oct-4, Nanog and Rex-1) and early hematopoietic stem cells (Scl, HoxB4 and GATA 2) markers in these cells, and (iii) induced phosphorylation of MAPK p42/44 and serine-threonine kinase AKT. Furthermore, molecular analysis revealed that ES-MV express Wnt-3 protein and are selectively highly enriched in mRNA for several pluripotent transcription factors as compared to parental ES cells. More important, this mRNA could be delivered by ES-MV to target cells and translated into the corresponding proteins. The biological effects of ES-MV were inhibited after heat inactivation or pretreatment with RNAse, indicating a major involvement of protein and mRNA components of ES-MV in the observed phenomena. We postulate that ES-MV may efficiently expand HPC by stimulating them with ES-MV expressed ligands (e.g., Wnt-3) as well as increase their pluripotency after horizontal transfer of ES-derived mRNA.


Assuntos
Comunicação Celular/genética , Estruturas da Membrana Celular/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/metabolismo , Proteínas Wnt/metabolismo , Animais , Antígenos Ly/metabolismo , Transporte Biológico/genética , Linhagem Celular , Estruturas da Membrana Celular/genética , Sobrevivência Celular/fisiologia , Citoplasma/metabolismo , Embrião de Mamíferos/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Técnicas In Vitro , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fosforilação , Células-Tronco Pluripotentes/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transfecção , Proteína Wnt3
18.
Leukemia ; 20(1): 18-28, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16270036

RESUMO

The concept that bone marrow (BM)-derived cells participate in neural regeneration remains highly controversial and the identity of the specific cell type(s) involved remains unknown. We recently reported that the BM contains a highly mobile population of CXCR4+ cells that express mRNA for various markers of early tissue-committed stem cells (TCSCs), including neural TCSCs. Here, we report that these cells not only express neural lineage markers (beta-III-tubulin, Nestin, NeuN, and GFAP), but more importantly form neurospheres in vitro. These neural TCSCs are present in significant amounts in BM harvested from young mice but their abundance and responsiveness to gradients of motomorphogens, such as SDF-1, HGF, and LIF, decreases with age. FACS analysis, combined with analysis of neural markers at the mRNA and protein levels, revealed that these cells reside in the nonhematopoietic CXCR4+/Sca-1+/lin-/CD45 BM mononuclear cell fraction. Neural TCSCs are mobilized into the peripheral-blood following stroke and chemoattracted to the damaged neural tissue in an SDF-1-CXCR4-, HGF-c-Met-, and LIF-LIF-R-dependent manner. Based on these data, we hypothesize that the postnatal BM harbors a nonhematopoietic population of cells that express markers of neural TCSCs that may account for the beneficial effects of BM-derived cells in neural regeneration.


Assuntos
Antígenos de Diferenciação/metabolismo , Células da Medula Óssea/citologia , Neurônios/fisiologia , Células-Tronco/citologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Células da Medula Óssea/fisiologia , Linhagem da Célula , Movimento Celular , Quimiocina CXCL12 , Quimiocinas CXC/biossíntese , Feminino , Fator de Crescimento de Hepatócito/biossíntese , Técnicas In Vitro , Interleucina-6/biossíntese , Fator Inibidor de Leucemia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , RNA Mensageiro/biossíntese , Receptores CXCR4/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia
19.
Leukemia ; 31(8): 1671-1677, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28502982

RESUMO

Evidence has accumulated that normal human and murine hematopoietic stem cells express several functional pituitary and gonadal sex hormones, and that, in fact, some sex hormones, such as androgens, have been employed for many years to stimulate hematopoiesis in patients with bone marrow aplasia. Interestingly, sex hormone receptors are also expressed by leukemic cell lines and blasts. In this review, I will discuss the emerging question of why hematopoietic cells express these receptors. A tempting hypothetical explanation for this phenomenon is that hematopoietic stem cells are related to subpopulation of migrating primordial germ cells. To support of this notion, the anatomical sites of origin of primitive and definitive hematopoiesis during embryonic development are tightly connected with the migratory route of primordial germ cells: from the proximal epiblast to the extraembryonic endoderm at the bottom of the yolk sac and then back to the embryo proper via the primitive streak to the aorta-gonado-mesonephros (AGM) region on the way to the genital ridges. The migration of these cells overlaps with the emergence of primitive hematopoiesis in the blood islands at the bottom of the yolk sac, and definitive hematopoiesis that occurs in hemogenic endothelium in the embryonic dorsal aorta in AGM region.


Assuntos
Desenvolvimento Embrionário/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Movimento Celular , Hormônios Esteroides Gonadais/fisiologia , Hematopoese , Humanos
20.
Leukemia ; 31(2): 446-458, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27451975

RESUMO

As a crucial arm of innate immunity, the complement cascade (ComC) is involved both in mobilization of normal hematopoietic stem/progenitor cells (HSPCs) from bone marrow (BM) into peripheral blood and in their homing to BM. Despite the fact that ComC cleavage fragments alone do not chemoattract normal HSPCs, we found that leukemia cell lines as well as clonogenic blasts from chronic myeloid leukemia and acute myeloid leukemia patients respond robustly to C3 and C5 cleavage fragments by chemotaxis and increased adhesion. This finding was supported by the detection of C3a and C5a receptors in cells from human malignant hematopoietic cell lines and patient blasts at the mRNA (reverse transcriptase-polymerase chain reaction) and protein level (fluorescence-activated cell sorting), and by the demonstration that these receptors respond to stimulation by C3a and C5a by phosphorylation of p42/44 and p38 mitogen-activated protein kinases (MAPK), and protein kinase B (PKB/AKT). We also found that inducible heme oxygenase 1 (HO-1) is a negative regulator of ComC-mediated trafficking of leukemic cells, and that stimulation of leukemic cells by C3 or C5 cleavage fragments activates p38 MAPK, which downregulates HO-1 expression, rendering cells more mobile. We conclude that activation of the ComC in leukemia/lymphoma patients (for example, as a result of accompanying infections) enhances the motility of malignant cells and contributes to their spread in a p38 MAPK-HO-1-dependent manner. Therefore, inhibition of p38 MAPK or upregulation of HO-1 by small-molecule modulators would have a beneficial effect on ameliorating cell migration-mediated expansion of leukemia/lymphoma cells when the ComC becomes activated.


Assuntos
Ativação do Complemento/imunologia , Regulação Leucêmica da Expressão Gênica , Heme Oxigenase-1/genética , Leucemia/genética , Leucemia/imunologia , Animais , Adesão Celular/genética , Adesão Celular/imunologia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Quimiotaxia/genética , Quimiotaxia/imunologia , Complemento C3/imunologia , Complemento C3/metabolismo , Complemento C5/imunologia , Complemento C5/metabolismo , Regulação para Baixo , Citometria de Fluxo , Técnicas de Inativação de Genes , Células-Tronco Hematopoéticas/metabolismo , Xenoenxertos , Humanos , Imunofenotipagem , Camundongos , Proteólise , RNA Interferente Pequeno/genética , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/metabolismo , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA