Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Immunity ; 47(4): 680-696.e8, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045900

RESUMO

The classical model of hematopoiesis established in the mouse postulates that lymphoid cells originate from a founder population of common lymphoid progenitors. Here, using a modeling approach in humanized mice, we showed that human lymphoid development stemmed from distinct populations of CD127- and CD127+ early lymphoid progenitors (ELPs). Combining molecular analyses with in vitro and in vivo functional assays, we demonstrated that CD127- and CD127+ ELPs emerged independently from lympho-mono-dendritic progenitors, responded differently to Notch1 signals, underwent divergent modes of lineage restriction, and displayed both common and specific differentiation potentials. Whereas CD127- ELPs comprised precursors of T cells, marginal zone B cells, and natural killer (NK) and innate lymphoid cells (ILCs), CD127+ ELPs supported production of all NK cell, ILC, and B cell populations but lacked T potential. On the basis of these results, we propose a "two-family" model of human lymphoid development that differs from the prevailing model of hematopoiesis.


Assuntos
Linfócitos B/metabolismo , Células Matadoras Naturais/metabolismo , Células Progenitoras Linfoides/metabolismo , Linfopoese/genética , Linfócitos T/metabolismo , Adolescente , Adulto , Animais , Linfócitos B/citologia , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade alfa de Receptor de Interleucina-7/genética , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Células Matadoras Naturais/citologia , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/transplante , Masculino , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Pessoa de Meia-Idade , Transplante de Células-Tronco , Linfócitos T/citologia , Transplante Heterólogo , Adulto Jovem
2.
Proc Natl Acad Sci U S A ; 116(13): 6298-6307, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30846549

RESUMO

Natural regulatory T cells (nTregs) ensure the control of self-tolerance and are currently used in clinical trials to alleviate autoimmune diseases and graft-versus-host disease after hematopoietic stem cell transfer. Based on CD39/CD26 markers, blood nTreg analysis revealed the presence of five different cell subsets, each representing a distinct stage of maturation. Ex vivo added microenvironmental factors, including IL-2, TGFß, and PGE2, direct the conversion from naive precursor to immature memory and finally from immature to mature memory cells, the latest being a no-return stage. Phenotypic and genetic characteristics of the subsets illustrate the structural parental maturation between subsets, which further correlates with the expression of regulatory factors. Regarding nTreg functional plasticity, both maturation stage and microenvironmental cytokines condition nTreg activities, which include blockade of autoreactive immune cells by cell-cell contact, Th17 and IL-10 Tr1-like activities, or activation of TCR-stimulating dendritic cell tolerization. Importantly, blood nTreg CD39/CD26 profile remained constant over a 2-y period in healthy persons but varied from person to person. Preliminary data on patients with autoimmune diseases or acute myelogenous leukemia illustrate the potential use of the nTreg CD39/CD26 profile as a blood biomarker to monitor chronic inflammatory diseases. Finally, we confirmed that naive conventional CD4 T cells, TCR-stimulated under a tolerogenic conditioned medium, could be ex vivo reprogrammed to FOXP3 lineage Tregs, and further found that these cells were exclusively committed to suppressive function under all microenvironmental contexts.


Assuntos
Microambiente Celular/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/fisiologia , Apirase/sangue , Doenças Autoimunes/sangue , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Dinoprostona/metabolismo , Dipeptidil Peptidase 4/sangue , Fatores de Transcrição Forkhead/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Leucemia Mieloide , Células Th17/imunologia , Fator de Crescimento Transformador beta/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 314(3): L360-L371, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29167125

RESUMO

Distal lung diseases, such as pulmonary fibrosis or acute lung injury, are commonly associated with local alveolar hypoxia that may be deleterious through the stimulation of alveolar epithelial cell (AEC) apoptosis. In various murine models of alveolar injury, administration of allogenic human mesenchymal stem cells (hMSCs) exerts an overall protective paracrine effect, limiting lung inflammation and fibrosis. However, the precise mechanisms on lung cells themselves remain poorly understood. Here, we investigated whether hMSC-conditioned medium (hMSC-CM) would protect AECs from hypoxia-induced apoptosis and explored the mechanisms involved in this cytoprotective effect. Exposure of rat primary AECs to hypoxia (1.5% O2 for 24 h) resulted in hypoxia-inducible factor (HIF)-1α protein stabilization, partly dependent on reactive oxygen species (ROS) accumulation, and in a twofold increase in AEC apoptosis that was prevented by the HIF inhibitor 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl-indazole and the antioxidant drug N-acetyl cysteine. Incubation of AECs with hMSC-CM significantly reduced hypoxia-induced apoptosis. hMSC-CM decreased HIF-1α protein expression, as well as ROS accumulation through an increase in antioxidant enzyme activities. Expression of Bnip3 and CHOP, two proapoptotic targets of HIF-1α and ROS pathways, respectively, was suppressed by hMSC-CM, while Bcl-2 expression was restored. The paracrine protective effect of hMSC was partly dependent on keratinocyte growth factor and hepatocyte growth factor secretion, preventing ROS and HIF-1α accumulation.


Assuntos
Células Epiteliais Alveolares/citologia , Apoptose , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/fisiopatologia , Células-Tronco Mesenquimais/citologia , Alvéolos Pulmonares/citologia , Espécies Reativas de Oxigênio/metabolismo , Células Epiteliais Alveolares/fisiologia , Animais , Células Cultivadas , Humanos , Masculino , Células-Tronco Mesenquimais/fisiologia , Alvéolos Pulmonares/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
4.
Transfusion ; 58(2): 520-531, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29277910

RESUMO

After 30 years of hematopoietic stem cell use for various indications, umbilical cord blood is considered as an established source of cells with marrow and postmobilization peripheral blood. The limited number of cells still remains a problematic element restricting their use, especially in adults who require to be grafted with a higher cell number. Improving the quality of harvested cord blood, at least in terms of volume and amount of cells, is essential to decrease the number of discarded units. In this review, we examine several variables related to parturient, pregnancy, labor, delivery, collection, the newborn, umbilical cord, and placenta. We aim to understand the biologic mechanisms that can impact cord blood quality. This knowledge will ultimately allow targeting donors, which could provide a rich graft and improve the efficiency of the collection.


Assuntos
Preservação de Sangue , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sangue Fetal , Aloenxertos , Humanos
5.
Haematologica ; 102(6): 976-983, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28302713

RESUMO

Efforts to implement family cord blood banking have been developed in the past decades for siblings requiring stem cell transplantation for conditions such as sickle cell disease. However, public banks are faced with challenging decisions about the units to be stored, discarded, or used for other endeavors. We report here 20 years of experience in family cord blood banking for sickle cell disease in two dedicated public banks. Participants were pregnant women who had a previous child diagnosed with homozygous sickle cell disease. Participation was voluntary and free of charge. All mothers underwent mandatory serological screening. Cord blood units were collected in different hospitals, but processed and stored in two public banks. A total of 338 units were stored for 302 families. Median recipient age was six years (11 months-15 years). Median collected volume and total nucleated cell count were 91 mL (range 23-230) and 8.6×108 (range 0.7-75×108), respectively. Microbial contamination was observed in 3.5% (n=12), positive hepatitis B serology in 25% (n=84), and homozygous sickle cell disease in 11% (n=37) of the collections. Forty-four units were HLA-identical to the intended recipient, and 28 units were released for transplantation either alone (n=23) or in combination with the bone marrow from the same donor (n=5), reflecting a utilization rate of 8%. Engraftment rate was 96% with 100% survival. Family cord blood banking yields good quality units for sibling transplantation. More comprehensive banking based on close collaboration among banks, clinical and transplant teams is recommended to optimize the use of these units.


Assuntos
Anemia Falciforme/terapia , Armazenamento de Sangue/métodos , Transplante de Células-Tronco de Sangue do Cordão Umbilical/normas , Família , Sangue Fetal/citologia , Adolescente , Adulto , Bancos de Sangue/normas , Criança , Pré-Escolar , Feminino , Sobrevivência de Enxerto , Histocompatibilidade , Humanos , Lactente , Masculino , Gravidez , Irmãos , Taxa de Sobrevida , Doadores de Tecidos , Adulto Jovem
6.
Am J Physiol Lung Cell Mol Physiol ; 310(5): L439-51, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26702148

RESUMO

Administration of bone marrow-derived human mesenchymal stem cells (hMSC) reduces lung inflammation, fibrosis, and mortality in animal models of lung injury, by a mechanism not completely understood. We investigated whether hMSC would prevent epithelial-mesenchymal transition (EMT) induced by hypoxia in primary rat alveolar epithelial cell (AEC). In AEC cultured on semipermeable filters, prolonged hypoxic exposure (1.5% O2 for up to 12 days) induced phenotypic changes consistent with EMT, i.e., a change in cell morphology, a decrease in transepithelial resistance (Rte) and in the expression of epithelial markers [zonula occludens-1 (ZO-1), E-cadherin, AQP-5, TTF-1], together with an increase in mesenchymal markers [vimentin, α-smooth muscle actin (α-SMA)]. Expression of transcription factors driving EMT such as SNAIL1, ZEB1, and TWIST1 increased after 2, 24, and 48 h of hypoxia, respectively. Hypoxia also induced TGF-ß1 mRNA expression and the secretion of active TGF-ß1 in apical medium, and the expression of connective tissue growth factor (CTGF), two inducers of EMT. Coculture of AEC with hMSC partially prevented the decrease in Rte and in ZO-1, E-cadherin, and TTF-1 expression, and the increase in vimentin expression induced by hypoxia. It also abolished the increase in TGF-ß1 expression and in TGF-ß1-induced genes ZEB1, TWIST1, and CTGF. Finally, incubation with human recombinant KGF at a concentration similar to what was measured in hMSC-conditioned media restored the expression of TTF-1 and prevented the increase in TWIST1, TGF-ß1, and CTGF in hypoxic AEC. Our results indicate that hMSC prevent hypoxia-induced alveolar EMT through the paracrine modulation of EMT signaling pathways and suggest that this effect is partly mediated by KGF.


Assuntos
Células Epiteliais Alveolares/metabolismo , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Células-Tronco Mesenquimais/metabolismo , Animais , Hipóxia Celular , Linhagem Celular , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Pulmão/metabolismo , Masculino , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
7.
Eur Heart J ; 36(30): 2011-7, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25990469

RESUMO

AIMS: Comparative studies suggest that stem cells committed to a cardiac lineage are more effective for improving heart function than those featuring an extra-cardiac phenotype. We have therefore developed a population of human embryonic stem cell (ESC)-derived cardiac progenitor cells. METHODS AND RESULTS: Undifferentiated human ESCs (I6 line) were amplified and cardiac-committed by exposure to bone morphogenetic protein-2 and a fibroblast growth factor receptor inhibitor. Cells responding to these cardio-instructive cues express the cardiac transcription factor Isl-1 and the stage-specific embryonic antigen SSEA-1 which was then used to purify them by immunomagnetic sorting. The Isl-1(+) SSEA-1(+) cells were then embedded into a fibrin scaffold which was surgically delivered onto the infarct area in a 68-year-old patient suffering from severe heart failure [New York Heart Association [NYHA] functional Class III; left ventricular ejection fraction (LVEF): 26%]. A coronary artery bypass was performed concomitantly in a non-infarcted area. The implanted cells featured a high degree of purity (99% were SSEA-1(+)), had lost the expression of Sox-2 and Nanog, taken as markers for pluripotency, and strongly expressed Isl-1. The intraoperative delivery of the patch was expeditious. The post-operative course was uncomplicated either. After 3 months, the patient is symptomatically improved (NYHA functional Class I; LVEF: 36%) and a new-onset contractility is echocardiographically evident in the previously akinetic cell/patch-treated, non-revascularized area. There have been no complications such as arrhythmias, tumour formation, or immunosuppression-related adverse events. CONCLUSION: This observation demonstrates the feasibility of generating a clinical-grade population of human ESC-derived cardiac progenitors and combining it within a tissue-engineered construct. While any conclusion pertaining to efficacy would be meaningless, the patient's functional outcome yet provides an encouraging hint. Beyond this case, the platform that has been set could be useful for generating different ESC-derived lineage-specific progenies.


Assuntos
Insuficiência Cardíaca/terapia , Células-Tronco Embrionárias Humanas/transplante , Feminino , Humanos , Pessoa de Meia-Idade , Isquemia Miocárdica/terapia , Alicerces Teciduais , Resultado do Tratamento , Disfunção Ventricular Esquerda/terapia
8.
Eur Heart J ; 36(12): 743-50, 2015 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24835485

RESUMO

AIM: There is now compelling evidence that cells committed to a cardiac lineage are most effective for improving the function of infarcted hearts. This has been confirmed by our pre-clinical studies entailing transplantation of human embryonic stem cell (hESC)-derived cardiac progenitors in rat and non-human primate models of myocardial infarction. These data have paved the way for a translational programme aimed at a phase I clinical trial. METHODS AND RESULTS: The main steps of this programme have included (i) the expansion of a clone of pluripotent hESC to generate a master cell bank under good manufacturing practice conditions (GMP); (ii) a growth factor-induced cardiac specification; (iii) the purification of committed cells by immunomagnetic sorting to yield a stage-specific embryonic antigen (SSEA)-1-positive cell population strongly expressing the early cardiac transcription factor Isl-1; (iv) the incorporation of these cells into a fibrin scaffold; (v) a safety assessment focused on the loss of teratoma-forming cells by in vitro (transcriptomics) and in vivo (cell injections in immunodeficient mice) measurements; (vi) an extensive cytogenetic and viral testing; and (vii) the characterization of the final cell product and its release criteria. The data collected throughout this process have led to approval by the French regulatory authorities for a first-in-man clinical trial of transplantation of these SSEA-1(+) progenitors in patients with severely impaired cardiac function. CONCLUSION: Although several facets of this manufacturing process still need to be improved, these data may yet provide a useful platform for the production of hESC-derived cardiac progenitor cells under safe and cost-effective GMP conditions.


Assuntos
Células-Tronco Embrionárias Humanas/transplante , Separação Imunomagnética/métodos , Bancos de Tecidos/organização & administração , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ensaios Clínicos Fase I como Assunto , Análise Citogenética , Estudos de Avaliação como Assunto , Humanos , Camundongos SCID , Miócitos Cardíacos/citologia , Miócitos Cardíacos/transplante , Preservação de Tecido/métodos , Alicerces Teciduais
9.
Biol Blood Marrow Transplant ; 21(3): 569-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25528387

RESUMO

The pathophysiology of acute graft-versus-host disease (GVHD) remains poorly understood in humans. Although T cell subsets have been identified to play a major role in disease initiation in rodents, clinical data on the effect of these different subsets are scarce and conflicting. To address this question, immunophenotyping analyses were performed on the graft in 210 patients. The onset of acute GVHD was retrospectively correlated with these subpopulations. In an adjusted analysis, only the absolute count of CD45lo/CD62Llo CD8(+) T cells (effector memory T cells) was significantly associated with the onset of grade 2 to 4 acute GVHD. Thus, in contrast to experimental data, we found that the number of effector memory but not of naïve T cells was associated with the onset of GVHD. These results should be kept in mind while clinical trials, which aim to deplete naïve T cells, are underway in several institutions.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Memória Imunológica , Transplante de Células-Tronco , Doença Aguda , Adolescente , Adulto , Idoso , Aloenxertos , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Criança , Pré-Escolar , Feminino , Seguimentos , Doença Enxerto-Hospedeiro/sangue , Doença Enxerto-Hospedeiro/patologia , Humanos , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade
10.
Mol Ther ; 22(1): 219-25, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23831596

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset autosomal dominant genetic disease mainly characterized by ptosis and dysphagia. We conducted a phase I/IIa clinical study (ClinicalTrials.gov NCT00773227) using autologous myoblast transplantation following myotomy in adult OPMD patients. This study included 12 patients with clinical diagnosis of OPMD, indication for cricopharyngeal myotomy, and confirmed genetic diagnosis. The feasibility and safety end points of both autologous myoblast transplantation and the surgical procedure were assessed by videoendoscopy in addition to physical examinations. Potential therapeutic benefit was also assessed through videoendoscopy and videofluoroscopy of swallowing, quality of life score, dysphagia grade, and a drink test. Patients were injected with a median of 178 million myoblasts following myotomy. Short and long-term (2 years) safety and tolerability were observed in all the patients, with no adverse effects. There was an improvement in the quality of life score for all 12 patients, and no functional degradation in swallowing was observed for 10 patients. A cell dose-dependant improvement in swallowing was even observed in this study. This trial supports the hypothesis that a local injection of autologous myoblasts in the pharyngeal muscles is a safe and efficient procedure for OPMD patients.


Assuntos
Distrofia Muscular Oculofaríngea/terapia , Mioblastos Esqueléticos/transplante , Idoso , Esfíncter Esofágico Superior/metabolismo , Esfíncter Esofágico Superior/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofia Muscular Oculofaríngea/diagnóstico , Distrofia Muscular Oculofaríngea/genética , Músculos Faríngeos/metabolismo , Músculos Faríngeos/fisiopatologia , Músculos Faríngeos/cirurgia , Transplante Autólogo , Resultado do Tratamento
11.
Am J Physiol Lung Cell Mol Physiol ; 306(11): L975-85, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24682451

RESUMO

Mesenchymal stromal cells (MSCs) or their media (MSC-M) were reported to reverse acute lung injury (ALI)-induced decrease of alveolar fluid clearance. To determine the mechanisms by which MSC-M exert their beneficial effects, an in vitro model of alveolar epithelial injury was created by exposing primary rat alveolar epithelial cells (AECs) to hypoxia (3% O2) plus cytomix, a combination of IL-1ß, TNF-α, and IFN-γ. MSC-M were collected from human MSCs exposed for 12 h to either normoxia (MSC-M) or to hypoxia plus cytomix (HCYT-MSC-M). This latter condition was used to model the effect of alveolar inflammation and hypoxia on paracrine secretion of MSCs in the injured lung. Comparison of paracrine soluble factors in MSC media showed that the IL-1 receptor antagonist and prostaglandin E2 were markedly increased while keratinocyte growth factor (KGF) was twofold lower in HCYT-MSC-M compared with MSC-M. In AECs, hypoxia plus cytomix increased protein permeability, reduced amiloride-sensitive short-circuit current (AS-Isc), and also decreased the number of α-epithelial sodium channel (α-ENaC) subunits in the apical membrane. To test the effects of MSC media, MSC-M and HCYT-MSC-M were added for an additional 12 h to AECs exposed to hypoxia plus cytomix. MSC-M and HCYT-MSC-M completely restored epithelial permeability to normal. MSC-M, but not HCYT-MSC-M, significantly prevented the hypoxia plus cytomix-induced decrease of ENaC activity and restored apical α-ENaC channels. Interestingly, KGF-deprived MSC-M were unable to restore amiloride-sensitive sodium transport, indicating a possible role for KGF in the beneficial effect of MSC-M. These results indicate that MSC-M may be a preferable therapeutic option for ALI.


Assuntos
Células Epiteliais Alveolares/metabolismo , Células-Tronco Mesenquimais/metabolismo , Sódio/metabolismo , Animais , Apoptose , Transporte Biológico , Hipóxia Celular , Permeabilidade da Membrana Celular , Células Cultivadas , Meios de Cultivo Condicionados , Dinoprostona/metabolismo , Canais Epiteliais de Sódio/metabolismo , Fator 7 de Crescimento de Fibroblastos/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Masculino , Comunicação Parácrina , Alvéolos Pulmonares/patologia , Ratos , Ratos Sprague-Dawley
12.
J Immunol ; 189(4): 1648-60, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22798679

RESUMO

To model the developmental pattern of human prothymocytes and thymopoiesis, we used NOD-scid/γc(-/-) mice grafted with human umbilical cord blood CD34(+) hematopoietic progenitor cells (HPCs). Human prothymocytes developed in the murine bone marrow (BM) from multipotent CD34(++)CD38(lo)lineage(-) HPCs to CD34(++)CD7(+)CD2(-) pro-T1 cells that progressed in a Notch-dependent manner to CD34(+)CD7(++)CD2(+) pro-T2 cells, which migrated to the thymus. BM prothymocyte numbers peaked 1 mo after graft, dropped at mo 2, and persisted at low levels thereafter, with only a few CD34(+)CD7(lo) prothymocytes with limited T potential being detected by mo 5. As a consequence, thymopoiesis in this xenogeneic setting began by weeks 4-6, peaked at mo 3, and decreased thenceforth. Analyzing mice grafted at 2, 4 or 8, mo of age showed that in an "older" BM, prothymocyte differentiation was perturbed and resulted in CD34(+)CD7(lo) prothymocytes with limited T potential. Whereas the early drop in BM thymopoietic activity was related to a Notch-independent loss of T potential by CD34(++)CD38(lo)lineage(-) HPCs, the later age-dependent production decline of prothymocytes was linked to a more complex mix of cell-intrinsic and microenvironmental defects. Accordingly, and contrasting with what was observed with umbilical cord blood HPCs, CD34(+) HPCs from human adult BM displayed only marginal thymopoietic activity when grafted into young 2-mo-old NOD-scid/γc(-/-) mice. These data demonstrate that the developmental pattern of BM prothymocytes during human late fetal and early postnatal life can be reproduced in humanized mice, and they suggest that onset of human thymus involution relates to decreased colonization by prothymocytes.


Assuntos
Diferenciação Celular/imunologia , Células Progenitoras Linfoides/citologia , Linfopoese/fisiologia , Linfócitos T/citologia , Timo/citologia , Animais , Células da Medula Óssea/citologia , Linhagem da Célula/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
13.
Angiogenesis ; 16(4): 821-36, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23748743

RESUMO

Circulating endothelial progenitor cells (cEPC) are capable of homing to neovascularisation sites, in which they proliferate and differentiate into endothelial cells. Transplantation of cEPC-derived cells, in particular those isolated from umbilical cord blood (UCB), has emerged as a promising approach in the treatment of cardio-vascular diseases. After in vivo transplantation, these cells may be exposed to local or systemic inflammation or pathogens, of which they are a common target. Because Toll-like receptors (TLR) are critical in detecting pathogens and in initiating inflammatory responses, we hypothesized that TLR may govern UCB cEPC-derived cells function. While these cells expressed almost all TLR, we found that only TLR3 dramatically impaired cell properties. TLR3 activation inhibited cell proliferation, modified cell cycle entry, impaired the in vitro angiogenic properties and induced pro-inflammatory cytokines production. The anti-angiogenic effect of TLR3 activation was confirmed in vivo in a hind-limb ischemic mice model. Moreover, TLR3 activation consistently leads to an upregulation of miR-29b, -146a and -155 and to a deregulation of cytoskeleton and cell cycle regulator. Hence, TLR3 activation is likely to be a key regulator of cEPC-derived cells properties.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica/fisiologia , Receptor 3 Toll-Like/fisiologia , Animais , Ciclo Celular , Divisão Celular , Movimento Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/genética , Células Endoteliais/citologia , Endotélio Vascular/fisiologia , Feminino , Sangue Fetal/citologia , Regulação da Expressão Gênica/fisiologia , Membro Posterior/irrigação sanguínea , Humanos , Recém-Nascido , Isquemia/cirurgia , Ligantes , Lipoproteínas LDL/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , MicroRNAs/genética , Oligonucleotídeos/farmacologia , Poli I-C/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/biossíntese , Receptores Toll-Like/agonistas , Receptores Toll-Like/biossíntese , Receptores Toll-Like/genética , Fator de Necrose Tumoral alfa/farmacologia , Cicatrização
14.
J Cell Mol Med ; 16(7): 1544-52, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21895965

RESUMO

Transplantation of allogeneic human embryonic stem cell-derived cardiac progenitors triggers an immune response. We assessed whether this response could be modulated by the concomitant use of adipose-derived stromal cells (ADSC). Peripheral blood mononuclear cells were collected from 40 patients with coronary artery disease (CAD) and nine healthy controls. Cardiac progenitors (CD15(+) Mesp1(+)) were generated as already reported from the I6 cell line treated with bone morphogenetic protein (BMP)-2. Adipose-derived stromal cells were obtained from abdominal dermolipectomies. We assessed the proliferative response of peripheral lymphocytes from patients and controls to cardiac progenitors cultured on a monolayer of ADSC, to allogeneic lymphocytes in mixed lymphocyte culture and to the T cell mitogen phytohemaglutin A in presence or absence of ADSC. Cardiac progenitors cultured on a monolayer of ADSC triggered a proliferation of lymphocytes from both patients and controls albeit lower than that induced by allogeneic lymphocytes. When cultured alone, ADSC did not induce any proliferation of allogeneic lymphocytes. When added to cultures of lymphocytes, ADSC significantly inhibited the alloantigen or mitogen-induced proliferative response. Compared to healthy controls, lymphocytes from patients presenting CAD expressed a decreased proliferative capacity, in particular to mitogen-induced stimulation. Adipose-derived stromal cells express an immunomodulatory effect that limits both alloantigen and mitogen-induced lymphocyte responses. Furthermore, lymphocytes from patients with CAD are low responders to conventional stimuli, possibly because of their age and disease-associated treatment regimens. We propose that, in combination, these factors may limit the in vivo immunogenicity of cardiac progenitors co-implanted with ADSC in patients with CAD.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Embrionárias/transplante , Leucócitos Mononucleares/citologia , Transplante de Células-Tronco/métodos , Células Estromais/citologia , Adipócitos/citologia , Adipócitos/imunologia , Adipócitos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Doença da Artéria Coronariana/fisiopatologia , Células-Tronco Embrionárias/citologia , Coração , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Pessoa de Meia-Idade , Mitógenos , Células Estromais/metabolismo , Adulto Jovem
15.
Circulation ; 122(11 Suppl): S118-23, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20837902

RESUMO

BACKGROUND: The safety and efficacy of myocardial regeneration using embryonic stem cells are limited by the risk of teratoma and the high rate of cell death. METHODS AND RESULTS: To address these issues, we developed a composite construct made of a sheet of adipose tissue-derived stroma cells and embryonic stem cell-derived cardiac progenitors. Ten Rhesus monkeys underwent a transient coronary artery occlusion followed, 2 weeks later, by the open-chest delivery of the composite cell sheet over the infarcted area or a sham operation. The sheet was made of adipose tissue-derived stroma cells grown from a biopsy of autologous adipose tissue and cultured onto temperature-responsive dishes. Allogeneic Rhesus embryonic stem cells were committed to a cardiac lineage and immunomagnetically sorted to yield SSEA-1(+) cardiac progenitors, which were then deposited onto the cell sheet. Cyclosporine was given for 2 months until the animals were euthanized. Preimplantation studies showed that the SSEA-1(+) progenitors expressed cardiac markers and had lost pluripotency. After 2 months, there was no teratoma in any of the 5 cell-treated monkeys. Analysis of >1500 histological sections showed that the SSEA-1(+) cardiac progenitors had differentiated into cardiomyocytes, as evidenced by immunofluorescence and real-time polymerase chain reaction. There were also a robust engraftment of autologous adipose tissue-derived stroma cells and increased angiogenesis compared with the sham animals. CONCLUSIONS: These data collected in a clinically relevant nonhuman primate model show that developmentally restricted SSEA-1(+) cardiac progenitors appear to be safe and highlight the benefit of the epicardial delivery of a construct harboring cells with a cardiomyogenic differentiation potential and cells providing them the necessary trophic support.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Embrionárias/transplante , Infarto do Miocárdio/terapia , Miocárdio/patologia , Regeneração , Transplante de Células-Tronco/métodos , Tecido Adiposo/transplante , Animais , Diferenciação Celular , Modelos Animais de Doenças , Humanos , Antígenos CD15 , Macaca mulatta , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , Células Estromais , Transplante Autólogo , Transplante Homólogo
16.
Br J Haematol ; 150(1): 93-101, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20507316

RESUMO

One of the cardinal symptoms of type 1 Gaucher Disease (GD) is cytopenia, usually explained by bone marrow (BM) infiltration by Gaucher cells and hypersplenism. However, some cases of cytopenia in splenectomized or treated patients suggest possible other mechanisms. To evaluate intra-cellular glucocerebrosidase (GlcC) activity in immature progenitors and to prove the conduritol B epoxide (CBE)-induced inhibition of the enzyme, we used an adapted flow cytometric technique before assessing the direct effect of GlcC deficiency in functional assays. Among haematopoietic cells from healthy donors, monocytes showed the highest GlcC activity but immature CD34(+) and mesenchymal cells also had significant GlcC activity. CBE greatly inhibited the enzyme activity of all cell categories. GlcC-deficient CD34(+) cells showed impaired ability to proliferate and differentiate in the expansion assay and had lower frequency of erythroid burst-forming units, granulocyte colony-forming units (CFU) and macrophage CFU progenitors, but the effect of GlcC deficiency on megakaryocyte CFU lineage was not significant. GlcC deficiency strongly impaired primitive haematopoiesis in long-term culture. Furthermore, GlcC deficiency progressively impaired proliferation of mesenchymal progenitors. These data suggest an intrinsic effect of GlcC deficiency on BM immature cells that supplements the pathophysiology of GD and opens new perspectives of therapeutic approach.


Assuntos
Doença de Gaucher/enzimologia , Glucosilceramidase/deficiência , Hematopoese/fisiologia , Células da Medula Óssea/enzimologia , Proliferação de Células , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo/métodos , Doença de Gaucher/fisiopatologia , Glucosilceramidase/antagonistas & inibidores , Glucosilceramidase/sangue , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Humanos , Inositol/análogos & derivados , Inositol/farmacologia , Modelos Biológicos , Técnicas de Cultura de Tecidos
17.
Exp Cell Res ; 315(19): 3396-405, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19615360

RESUMO

UNLABELLED: The success of hepatocyte transplantation has been limited by the low efficiency of transplanted cell integration into liver parenchyma. Human fetal hepatic progenitor cells (hepatoblasts) engraft more effectively than adult hepatocytes in mouse livers. However, the signals required for their integration are not yet fully understood. We investigated the role of HGF on the migration and invasive ability of human hepatic progenitors in vitro and in vivo. Hepatoblasts were isolated from the livers of human fetuses between 10 and 12 weeks of gestation. Their invasive ability was assessed in the presence or absence of HGF. These cells were also transplanted into immunodeficient mice and analyzed by immunohistochemistry. In contrast to TNF-alpha, HGF increased the motogenesis and invasiveness of hepatoblasts, but not of human adult hepatocytes, via phosphorylation of extracellular signal-regulated kinase (ERK) 1/2. The invasive ability of human hepatoblasts correlated with the expression and secretion of matrix metalloproteinases (MMPs). Hepatoblasts stimulated with HGF prior transplantation into newborn mice migrated from the portal area into the hepatic parenchyma. CONCLUSIONS: In contrast to adult hepatocytes, hepatoblasts display invasive ability that can be modulated by HGF in vitro and in vivo.


Assuntos
Movimento Celular , Fator de Crescimento de Hepatócito/fisiologia , Hepatócitos/transplante , Transplante de Células-Tronco/métodos , Células-Tronco/fisiologia , Animais , Proliferação de Células , Feto , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Camundongos , Camundongos SCID , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Células-Tronco/citologia , Transplante Heterólogo
18.
Br J Haematol ; 145(1): 107-14, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19183190

RESUMO

The potential role of the infused B cell subset after Hematopoietic Stem Cell Transplantation has not been yet studied. The present study analyzed the impact of B cells on transplant outcome in 254 patients who received a bone marrow graft from a human leucocyte antigen-identical sibling donor. The influence of B lineage-specific hematopoietic progenitor cells (CD34(+) CD19(+)) and B cells (immature and mature B cells, CD34(-) CD19(+)) was also analyzed. All included patients received a myeloablative regimen. The cumulative incidence function of acute graft-versus-host (GvHD) grade II to IV was 48% and was inversely associated with the number of CD34(+) CD19(+). There were no statistically significant associations between B cell subsets and chronic GvHD or survival. The CD34(+) CD19(+) B cell subset remained significantly associated with acute GvHD in multivariate analysis (Relative risk = 0.32, 95% confidence interval: 0.11-0.92, P = 0.035). In conclusion, a higher B lineage-specific hematopoietic progenitor cells (CD34(+) CD19(+)) cell dose is associated with a significant decrease incidence of acute GvHD.


Assuntos
Antígenos CD19/imunologia , Antígenos CD34/imunologia , Linfócitos B/imunologia , Transplante de Medula Óssea/imunologia , Adolescente , Adulto , Doenças da Medula Óssea/cirurgia , Feminino , Citometria de Fluxo/métodos , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/imunologia , Humanos , Masculino , Análise Multivariada , Modelos de Riscos Proporcionais , Risco , Irmãos , Células-Tronco/imunologia , Taxa de Sobrevida , Transplante Isogênico
19.
Bull Acad Natl Med ; 193(3): 537-43; discussion 543-4, 2009 Mar.
Artigo em Francês | MEDLINE | ID: mdl-19883008

RESUMO

The discovery of circulating endothelial progenitor cells (EPCs) in adult peripheral blood has opened up many exciting possibilities in vascular biology. Several studies have confirmed the existence of EPCs, as well as their bone marrow origin and their ability to integrate into vascular structures at sites of neoangiogenesis. EPCs appear to be naturally involved in the prevention of ischemia by participating directly in the vascularization process. Given their tropism for sites of neoangiogenesis, EPCs have clear therapeutic potential for treating ischemic diseases. If associated with other cell therapy products, they could improve tissue regeneration by promoting graft vascularization. However, the use of EPCs as a cell therapy product is limited by their rarity in peripheral blood. Cord blood contains many more EPCs, which are functional and can be expanded in culture. Their clinical use will require expansion in strictly controlled conditions and rigorous validation in preclinical models. EPCs could also serve as a quality markerforfrozen cord blood, showing the presence of non hematopoietic stem cells.


Assuntos
Doenças Cardiovasculares/terapia , Células Endoteliais/fisiologia , Cordão Umbilical/citologia , Células Endoteliais/transplante , Humanos , Neovascularização Fisiológica , Transplante de Células-Tronco
20.
J Tissue Eng Regen Med ; 13(12): 2191-2203, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31670903

RESUMO

In pathologies of the esophagus such as esophageal atresia, cancers, and caustic injuries, methods for full thickness esophageal replacement require the sacrifice of healthy intra-abdominal organs such as the stomach and the colon and are associated with high morbidity, mortality, and poor functional results. To overcome these problems, tissue engineering methods are developed to create a substitute with scaffolds and cells. The aim of this study was to develop a simple and safe decellularization process in order to obtain a clinical grade esophageal extracellular matrix. Following the decontamination step, porcine esophagi were decellularized in a bioreactor with sodium dodecyl sulfate and ethylenediaminetetraacetic acid for 3 days and were rinsed with deionized water. DNA was eliminated by a 3-hr DNase treatment. To remove any residual detergent, the matrix was then incubated with an absorbing resin. The resulting porcine esophageal matrix was characterized by the assessment of the efficiency of the decellularization process (DNA quantification), evaluation of sterility and absence of cytotoxicity, and its composition and biomechanical properties, as well as the possibility to be reseeded with mesenchymal stem cells. Complete decellularization with the preservation of the general structure, composition, and biomechanical properties of the native esophageal matrix was obtained. Sterility was maintained throughout the process, and the matrix showed no cytotoxicity. The resulting matrix met clinical grade criteria and was successfully reseeded with mesenchymal stem cells..


Assuntos
Esôfago/química , Matriz Extracelular/química , Teste de Materiais , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Células-Tronco Mesenquimais/citologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA