Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Stem Cell Rev Rep ; 16(3): 524-540, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32020407

RESUMEN

Nongonadal tissues express luteinizing hormone-chorionic gonadotropin receptors (LHCG-R) which are essential for their growth during fetal development. Adult mesenchymal stem/stromal cells (MSCs) have been shown to express functional LHCG-R outside pregnancy conditions, making them susceptible to hCG stimulation. In the present study we tested the effect of hCG treatment on bone marrow (BM) derived adherent stem cells in vitro, isolated from a parous women, mother of male sons, in order to evaluate its effect on maternal MSCs and in the same time on fetal microchimeric stem cells (FMSCs), to better understand the outcomes of this safe and affordable treatment on cell proliferation and expression of pluripotency genes. Our study highlights the beneficial effects of hCG exposure on gene regulation in bone marrow adherent stem cells through the upregulation of pluripotency genes and selection of more primitive mesenchymal stem cells with a better differentiation potential. Validation of these effects on MSCs and FMSCs long after parturition in vivo represents a close perspective as it could set the premises of a new mobilization strategy for the stem cell transplantation procedures in the clinical setting.


Asunto(s)
Células de la Médula Ósea/citología , Quimerismo/efectos de los fármacos , Gonadotropina Coriónica/farmacología , Células Madre Fetales/citología , Células Madre Fetales/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Regeneración/efectos de los fármacos , Adipocitos/citología , Adipocitos/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Separación Celular , Condrocitos/citología , Condrocitos/efectos de los fármacos , Femenino , Células Madre Fetales/efectos de los fármacos , Células Madre Fetales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética
3.
BMC Res Notes ; 11(1): 343, 2018 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-29843819

RESUMEN

OBJECTIVE: Human amniotic epithelial cells (hAECs) are a novel source of stem cells and have immunomodulatory effects on both the innate and adoptive immune system. hAECs can differentiate into multiple cell lineages that make them a suitable cell source for regenerative medicine. These cells express multiple toll-like receptors (TLRs) and respond to various TLR ligands. This study aimed to evaluate the effect of lipopolysaccharide (LPS), a TLR4 ligand, on the level of immunomodulatory and immunostimulatory factors of hAECs. RESULTS: Our results indicated that LPS had the ability to up-regulate the expression of prostaglandin E2 synthase and transforming growth factor-beta1 in hAECs. However, there was no change in the level of interleukin-1beta, interleukin-6 and interleukin-10 in hAECs when were stimulated with LPS. In addition, we observed tumor necrosis factor-alpha was only expressed at very low level in some of hAECs samples which its expression was independent of the effects of LPS.


Asunto(s)
Amnios/citología , Citocinas/efectos de los fármacos , Células Epiteliales , Células Madre Fetales , Factores Inmunológicos/farmacología , Lipopolisacáridos/farmacología , Prostaglandina-E Sintasas/efectos de los fármacos , Adulto , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Células Madre Fetales/efectos de los fármacos , Células Madre Fetales/inmunología , Células Madre Fetales/metabolismo , Humanos , Embarazo
4.
Stem Cell Reports ; 6(1): 137-49, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26724903

RESUMEN

B cells are key components of cellular and humoral immunity and, like all lymphocytes, are thought to originate and renew from hematopoietic stem cells (HSCs). However, our recent single-HSC transfer studies demonstrate that adult bone marrow HSCs do not regenerate B-1a, a subset of tissue B cells required for protection against pneumonia, influenza, and other infections. Since B-1a are regenerated by transfers of fetal liver, the question arises as to whether B-1a derive from fetal, but not adult, HSCs. Here we show that, similar to adult HSCs, fetal HSCs selectively fail to regenerate B-1a. We also show that, in humanized mice, human fetal liver regenerates tissue B cells that are phenotypically similar to murine B-1a, raising the question of whether human HSC transplantation, the mainstay of such models, is sufficient to regenerate human B-1a. Thus, our studies overtly challenge the current paradigm that HSCs give rise to all components of the immune system.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Células Madre Fetales/trasplante , Trasplante de Tejido Fetal/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Animales Recién Nacidos , Linfocitos B/citología , Linfocitos B/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Células Madre Fetales/inmunología , Células Madre Fetales/metabolismo , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Hígado/citología , Hígado/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos
5.
PLoS One ; 10(5): e0125414, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25933067

RESUMEN

Cell number in the mouse thymus increases steadily during the first two weeks after birth. It then plateaus and begins to decline by seven weeks after birth. The factors governing these dramatic changes in cell production are not well understood. The data herein correlate levels of High mobility group A 2 protein (Hmga2) expression with these temporal changes in thymopoiesis. Hmga2 is expressed at high levels in murine fetal and neonatal early T cell progenitors (ETP), which are the most immature intrathymic precursors, and becomes almost undetectable in these progenitors after 5 weeks of age. Hmga2 expression is critical for the initial, exponential expansion of thymopoiesis, as Hmga2 deficient mice have a deficit of ETPs within days after birth, and total thymocyte number is repressed compared to wild type littermates. Finally, our data raise the possibility that similar events occur in humans, because Hmga2 expression is high in human fetal thymic progenitors and falls in these cells during early infancy.


Asunto(s)
Envejecimiento/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Fetales/metabolismo , Proteína HMGA2/genética , Linfocitos T/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Envejecimiento/inmunología , Animales , Animales Recién Nacidos , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD/metabolismo , Diferenciación Celular , Proliferación Celular , Embrión de Mamíferos , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Células Madre Fetales/citología , Células Madre Fetales/inmunología , Feto , Regulación del Desarrollo de la Expresión Génica , Proteína HMGA2/inmunología , Proteína HMGA2/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Organogénesis/genética , Organogénesis/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Timocitos/citología , Timocitos/inmunología , Timo/citología , Timo/embriología
6.
Placenta ; 36(4): 410-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25684656

RESUMEN

INTRODUCTION: Gestational diabetes (GDM) is associated with long-term cardiovascular and metabolic diseases in offspring. However, the mechanisms are not well understood. We explored whether fetal exposure to a diabetic environment is associated with fetal endothelial progenitor cell dysfunction, and whether vitamin D can reverse the impairment. METHODS: Nineteen women with uncomplicated pregnancies and 18 women with GDM were recruited before delivery. Time to first appearance of endothelial colony forming cell (ECFC) colonies and number of ECFC colonies formed from culture of cord peripheral blood mononuclear cells were determined. Angiogenesis-related functions of ECFCs in vitro were tested in the presence or absence of vitamin D. RESULTS: Fetal ECFCs from GDM pregnancies formed fewer colonies in culture (P = 0.04) and displayed reduced proliferation (P = 0.02), migration (P = 0.04) and tubule formation (P = 0.03) compared to uncomplicated pregnancies. Fetal ECFCs exposed to hyperglycemia in vitro exhibited less migration (P < 0.05) and less tubule formation (P < 0.05) than normoglycemic control. Vitamin D significantly improved the dysfunction of fetal ECFCs from pregnancies complicated by GDM or after exposure of healthy ECFCs to hyperglycemia. DISCUSSION: Fetal ECFCs from GDM pregnancies or ECFCs exposed to hyperglycemia in vitro exhibit reduced quantity and impaired angiogenesis-related functions. Vitamin D significantly rescues these functions. These findings may have implications for vascular function of infants exposed to a diabetic intrauterine environment.


Asunto(s)
Calcitriol/metabolismo , Diabetes Gestacional/metabolismo , Angiopatías Diabéticas/etiología , Endotelio Vascular/metabolismo , Células Madre Fetales/metabolismo , Neovascularización Patológica/etiología , Vasculitis Sistémica/etiología , Adulto , Movimiento Celular , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Diabetes Gestacional/inmunología , Diabetes Gestacional/patología , Diabetes Gestacional/fisiopatología , Angiopatías Diabéticas/prevención & control , Suplementos Dietéticos , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Femenino , Sangre Fetal , Células Madre Fetales/inmunología , Células Madre Fetales/patología , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Neovascularización Patológica/prevención & control , Embarazo , Interferencia de ARN , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/antagonistas & inhibidores , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Estudios Retrospectivos , Vasculitis Sistémica/prevención & control , Vitamina D/metabolismo , Vitamina D/uso terapéutico
7.
J Hepatol ; 61(5): 1097-105, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24953023

RESUMEN

BACKGROUND & AIMS: Human biliary tree stem/progenitor cells (hBTSCs) are multipotent epithelial stem cells, easily obtained from the biliary tree, with the potential for regenerative medicine in liver, biliary tree, and pancreas diseases. Recent reports indicate that human mesenchymal stem cells are able to modulate the T cell immune response. However, no information exists on the capabilities of hBTSCs to control the allogeneic response. The aims of this study were to evaluate FasL expression in hBTSCs, to study the in vitro interaction between hBTSCs and human lymphocytes, and the role of Fas/FasL modulation in inducing T cell apoptosis in hBTSCs/T cell co-cultures. METHODS: Fas and FasL expression were evaluated in situ and in vitro by immunofluorescence and western blotting. Co-cultures of hBTSCs with human leukocytes were used to analyze the influence of hBTSCs on lymphocytes activation and apoptosis. RESULTS: hBTSCs expressed HLA antigens and FasL in situ and in vitro. Western blot data demonstrated that hBTSCs constitutively expressed high levels of FasL that increased after co-culture with T cells. Confocal microscopy demonstrated that FasL expression was restricted to EpCAM(+)/LGR5(+) cells. FACS analysis of T cells co-cultured with hBTSCs indicated that hBTSCs were able to induce apoptosis in activated CD4(+) and CD8(+) T cell populations. Moreover, the Fas receptor appears to be more expressed in T cells co-cultured with hBTSCs than in resting T cells. CONCLUSIONS: Our data suggest that hBTSCs could modulate the T cell response through the production of FasL, which influences the lymphocyte Fas/FasL pathway by inducing "premature" apoptosis in CD4(+) and CD8(+) T cells.


Asunto(s)
Sistema Biliar/citología , Sistema Biliar/inmunología , Proteína Ligando Fas/metabolismo , Células Madre Multipotentes/citología , Células Madre Multipotentes/inmunología , Receptor fas/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/inmunología , Células Madre Adultas/metabolismo , Apoptosis/inmunología , Sistema Biliar/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Técnicas de Cocultivo , Células Madre Fetales/citología , Células Madre Fetales/inmunología , Células Madre Fetales/metabolismo , Humanos , Inmunomodulación , Activación de Linfocitos , Células Madre Multipotentes/metabolismo , Transducción de Señal
8.
Blood ; 124(6): 973-80, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24879814

RESUMEN

Although in utero hematopoietic cell transplantation is a promising strategy to treat congenital hematopoietic disorders, levels of engraftment have not been therapeutic for diseases in which donor cells have no survival advantage. We used an antibody against the murine c-Kit receptor (ACK2) to deplete fetal host hematopoietic stem cells (HSCs) and increase space within the hematopoietic niche for donor cell engraftment. Fetal mice were injected with ACK2 on embryonic days 13.5 to 14.5 and surviving pups were transplanted with congenic hematopoietic cells on day of life 1. Low-dose ACK2 treatment effectively depleted HSCs within the bone marrow with minimal toxicity and the antibody was cleared from the serum before the neonatal transplantation. Chimerism levels were significantly higher in treated pups than in controls; both myeloid and lymphoid cell chimerism increased because of higher engraftment of HSCs in the bone marrow. To test the strategy of repeated HSC depletion and transplantation, some mice were treated with ACK2 postnatally, but the increase in engraftment was lower than that seen with prenatal treatment. We demonstrate a successful fetal conditioning strategy associated with minimal toxicity. Such strategies could be used to achieve clinically relevant levels of engraftment to treat congenital stem cell disorders.


Asunto(s)
Células Madre Fetales/citología , Terapias Fetales/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Acondicionamiento Pretrasplante/métodos , Animales , Animales Recién Nacidos , Femenino , Células Madre Fetales/inmunología , Células Madre Hematopoyéticas/inmunología , Ratones , Ratones Endogámicos C57BL , Embarazo , Proteínas Proto-Oncogénicas c-kit/administración & dosificación , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/inmunología , Nicho de Células Madre/inmunología
9.
Clin Immunol ; 152(1-2): 68-76, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24607604

RESUMEN

RATIONALE: Cord blood eosinophil/basophil progenitor cells (Eo/B) of high risk infants have been shown to predict respiratory illnesses in infancy. Here we investigated this association in a population-based cohort. Furthermore, we analysed whether newborns Th1/Th2 balance and prenatal environmental exposure impact Eo/B recruitment. METHODS: In a sub-cohort of the LINA study cord blood mononuclear cells were used for methylcellulose assays to assess Eo/B differentiation. Questionnaires were recorded during pregnancy and annually thereafter. Volatile organic compounds were measured during pregnancy and cord blood cytokines after ex vivo stimulation. RESULTS: Cord blood IL-4 and IL-13 positively correlated with Eo/B. Tobacco smoke related benzene was also positively associated with Eo/B. Enhanced Eo/B numbers increased the risk for wheezing within the first 24 months. CONCLUSIONS: The association between cord blood Eo/B and respiratory illnesses is not restricted to high-risk children. Prenatal environmental exposure and a Th2 milieu at birth contribute to Eo/B recruitment.


Asunto(s)
Basófilos/inmunología , Eosinófilos/inmunología , Células Madre Fetales/inmunología , Infecciones del Sistema Respiratorio/inmunología , Compuestos Orgánicos Volátiles/efectos adversos , Basófilos/citología , Basófilos/efectos de los fármacos , Diferenciación Celular , Estudios de Cohortes , Exposición a Riesgos Ambientales , Eosinófilos/citología , Eosinófilos/efectos de los fármacos , Femenino , Sangre Fetal/citología , Células Madre Fetales/citología , Células Madre Fetales/efectos de los fármacos , Humanos , Lactante , Recién Nacido , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/inmunología , Infecciones del Sistema Respiratorio/genética , Encuestas y Cuestionarios , Balance Th1 - Th2
10.
Fiziol Zh (1994) ; 59(4): 40-50, 2013.
Artículo en Ucraniano | MEDLINE | ID: mdl-24175475

RESUMEN

The aim of the study was to compare the mice-recipient's brain tissue cell-structural reactions in response to intracerebral implantation of syngeneic and allogeneic cell suspensions of neural progenitor cells (NPC) (E13-15). The NPC suspensions from mice-donors of C57BL/6 and CBA containing 72.7 +/- 9.9% Vimentin+ and 81, 812, 5% GFAP+ cells were inoculated by standard procedure in right temporal segment of cerebral hemisphere of mice-recipients C57BL/6 (1 x 10(6) cells per animal). The certain part of mice-recipients of allogeneic NPC were immunosupressed by Sandimmune (100 mkg per animal) on day 0, 3, 6 after neurotransplantation. The standard histological preparations of mice brains were performed after 24 hours, 6, 12, 18 and 37 days after NPC neurotransplantation, which were investigated by cytoanalyzer "IBAS" (Germany). After intracerebral inoculation of allogeneic foetal NPC the signs of the pericellular edema and lymphocyte infiltration were detected in adjacent brain sections on day 12-18 and decreased on day 37. Allogeneic foetal NPC were reserved till day 18 and revealed the signs of primary differentiation. After immunosupression by "Sandimmune" the foetal NPC underwent the phoenotypic differentiation and infiltration in related brain sections. On the day 37 the implanted NPC were not detected. Focal reaction of the brain glial component to implanted NPC declined faster after syngeneic NPC neuroimplantation (up to day 18) than after allogeneic NPC neuroimplantation (up to day 37). After the syngeneic NPC inoculation on the 37th day at the site of implantation the formation of a small fragment of immature bone was fixed, which may indicate the possibility of NPC transdifferentiation in other cell types.


Asunto(s)
Trasplante de Tejido Encefálico , Encéfalo/patología , Células Madre Fetales/trasplante , Supervivencia de Injerto/fisiología , Células-Madre Neurales/trasplante , Animales , Biomarcadores/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Diferenciación Celular , Transdiferenciación Celular , Ciclosporina/farmacología , Células Madre Fetales/citología , Células Madre Fetales/inmunología , Feto , Proteína Ácida Fibrilar de la Glía , Terapia de Inmunosupresión , Inmunosupresores/farmacología , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/inmunología , Trasplante Homólogo , Trasplante Isogénico , Vimentina/metabolismo
11.
J Neuroimmune Pharmacol ; 8(4): 1010-26, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23744346

RESUMEN

HIV enters the brain early during infection and induces a chronic inflammatory state that can result in neurological abnormalities in a subset of infected individuals. To investigate the effects of HIV exposure on neurogenesis and neuronal survival in the brain, we have used a model system consisting of human neuroepithelial progenitor (NEP) cells that undergo directed differentiation into astrocytes and neurons in vitro. Changes in gene expression in NEP cultures as a result of HIV exposure were investigated using gene expression microarrays with the Illumina HT-12 V4_0_R1 platform array. Through this approach, we identified a group of genes specifically upregulated by exposure to virus that are strongly related to interferon induced responses and antigen presentation. When the data were stratified by their apolipoprotein genotype, this innate immune response was more robust in the apolipoprotein E3/E3 genotype cultures than in the apolipoprotein E3/E4 counterparts. Biological processes as defined by the gene ontology (GO) program were also differently affected upon virus exposure in cultures of the two genotypes, particularly those related to antigen presentation and the actions of interferons. Differences occurred in both in numbers of genes affected and their significance in the GO processes in which they participate, with apoE3/E3 > apoE3/E4. These data suggest that maturing NEP cultures recognize HIV and respond to it by mounting an innate immune response with a vigor that is influenced by the apolipoprotein E genotype of the cells.


Asunto(s)
Apolipoproteínas E/fisiología , Células Madre Fetales/microbiología , VIH-1 , Inmunidad Innata/inmunología , Células-Madre Neurales/inmunología , Células Neuroepiteliales/inmunología , Apolipoproteínas E/genética , Células Cultivadas , Células Madre Fetales/inmunología , Células Madre Fetales/virología , Genotipo , Humanos , Inmunidad Innata/genética , Células-Madre Neurales/virología , Células Neuroepiteliales/virología
12.
Transplant Proc ; 44(8): 2490-4, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23026627

RESUMEN

BACKGROUND: Chronic allograft vasculopathy (CAV) is an important cause of graft loss. Considering the immune inflammatory events involved in the development of CAV, therapeutic approaches to target this process are of relevance. Human amniotic fluid-derived stem cells (hAFSCs), a class of fetal, pluripotent stem cells with intermediate characteristics between embryonic and adult stem cells, display immunomodulatory properties. hAFSCs express mesenchymal and embryonic markers, show high proliferation rates; however, they do not induce tumor formation, and their use does not raise ethical issues. Thus, we sought to investigate the effect of hAFSC on CAV in a model of aorta transplantation. METHODS: Orthotopic aorta transplantation was performed using Fisher (F344) rats as donors and Lewis rats as recipients. Rats were divided into three groups: syngeneic (SYNG), untreated F344 receiving aorta from F344 (n = 8); allogeneic (ALLO), Lewis rats receiving allogeneic aorta from F344 (n = 8); and ALLO + hAFSC, ALLO rats treated with hAFSC (10(6) cells; n = 8). Histological analysis and immunohistochemistry were performed 30 days posttransplantation. RESULTS: The ALLO group developed a robust aortic neointimal formation (208.7 ± 25.4 µm) accompanied by a significant high number of ED1+ (4845 ± 841 cells/mm2) and CD43+ cells (4064 ± 563 cells/mm2), and enhanced expression of α-smooth muscle actin in the neointima (25 ± 6%). Treatment with hAFSC diminished neointimal thickness (180.7 ± 23.7 µm) and induced a significant decrease of ED1+ (1100 ± 276 cells/mm2), CD43+ cells (1080 ± 309 cells/µm2), and α-smooth muscle actin expression 8 ± 3% in the neointima. CONCLUSIONS: These preliminary results showed that hAFSC suppressed inflammation and myofibroblast migration to the intima, which may contribute to ameliorate vascular changes in CAV.


Asunto(s)
Líquido Amniótico/citología , Aorta Abdominal/trasplante , Enfermedades de la Aorta/prevención & control , Células Madre Fetales/trasplante , Trasplante de Órganos/efectos adversos , Células Madre Pluripotentes/trasplante , Actinas/metabolismo , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Biomarcadores/metabolismo , Movimiento Celular , Células Cultivadas , Células Madre Fetales/inmunología , Células Madre Fetales/metabolismo , Humanos , Inmunohistoquímica , Masculino , Miofibroblastos/metabolismo , Miofibroblastos/patología , Neointima , Células Madre Pluripotentes/inmunología , Células Madre Pluripotentes/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Factores de Tiempo
13.
Stem Cells Dev ; 21(15): 2809-16, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-22731493

RESUMEN

Throughout every pregnancy, genetically distinct fetal microchimeric stem/progenitor cells (FMCs) engraft in the mother, persist long after delivery, and may home to damaged maternal tissues. Phenotypically normal fetal lymphoid progenitors have been described to develop in immunodeficient mothers in a fetus-treats-its-mother paradigm. Since stem cells contribute to muscle repair, we assessed this paradigm in the mdx mouse model of Duchenne muscular dystrophy. mdx females were bred serially to either ROSAeGFP males or mdx males to obtain postpartum microchimeras that received either wild-type FMCs or dystrophin-deficient FMCs through serial gestations. To enhance regeneration, notexin was injected into the tibialis anterior of postpartum mice. FMCs were detected by qPCR at a higher frequency in injected compared to noninjected side muscle (P=0.02). However, the number of dystrophin-positive fibers was similar in mothers delivering wild-type compared to mdx pups. In addition, there was no correlation between FMC detection and percentage dystrophin, and no GFP+ve FMCs were identified that expressed dystrophin. In 10/11 animals, GFP+ve FMCs were detected by immunohistochemistry, of which 60% expressed CD45 with 96% outside the basal lamina defining myofiber contours. Finally we confirmed lack of FMC contribution to statellite cells in postpartum mdx females mated with Myf5-LacZ males. We conclude that the FMC contribution to regenerating muscles is insufficient to have a functional impact.


Asunto(s)
Distrofina/deficiencia , Células Madre Fetales/fisiología , Músculo Esquelético/fisiopatología , Regeneración , Animales , Distrofina/biosíntesis , Distrofina/genética , Venenos Elapídicos/farmacología , Femenino , Células Madre Fetales/inmunología , Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Embarazo
14.
Trends Immunol ; 32(10): 493-503, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21802990

RESUMEN

Migration of hematopoietic stem cells (HSCs) is essential during embryonic development and throughout adult life. During embryogenesis, trafficking of HSCs is responsible for the sequential colonization of different hematopoietic organs by blood-producing cells. In adulthood, circulation of HSCs maintains homeostasis of the hematopoietic system and participates in innate immune responses. HSC trafficking is also crucial in clinical settings such as bone marrow (BM) and stem cell transplantation. This review provides an overview of the molecular and cellular signals that control and fine-tune trafficking of HSCs and hematopoietic progenitor cells in embryogenesis and during postnatal life. We also discuss the potential clinical utility of therapeutic approaches to modulate HSC trafficking in patients.


Asunto(s)
Células Madre Adultas/inmunología , Células de la Médula Ósea/inmunología , Movimiento Celular/inmunología , Células Madre Fetales/inmunología , Células Madre Hematopoyéticas/inmunología , Homeostasis/inmunología , Inmunidad Innata , Adulto , Células Madre Adultas/citología , Animales , Células de la Médula Ósea/citología , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Rastreo Celular , Embrión de Mamíferos , Desarrollo Embrionario/inmunología , Femenino , Células Madre Fetales/citología , Feto , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Humanos , Técnicas Inmunológicas , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Complejo GPIb-IX de Glicoproteína Plaquetaria , Embarazo
15.
J Vasc Res ; 47(2): 157-67, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19851077

RESUMEN

Inflammatory activation of the vascular endothelium is a major contributory factor to ischemic cardiovascular disease. Endothelial progenitor cells (EPCs) are being investigated for the treatment of ischemic disease or to coat vein grafts for bypass surgery. As an inflammatory environment might reduce their therapeutic efficacy, we sought to generate EPCs that are less sensitive to inflammatory activation. EPCs were obtained from human umbilical cord blood and transduced with a lentiviral vector for stable expression of A20, an anti-inflammatory protein. Nontransduced and green-fluorescent-protein-transduced cells were used as controls. Expression of A20 by EPCs did not modify cell morphology or expression of a panel of 20 proteins known to contribute to angiogenesis. Also, A20 had no effect on the capacity of EPCs to form tube-like structures in Matrigel. A20 expression reduced EPC activation by tumor necrosis factor-alpha and interleukin-1beta as determined from changes in vascular cell adhesion molecule 1 and E-selectin expression and decreased monocyte transmigration through a monolayer of EPCs. In conclusion, EPCs can be genetically modified to overexpress A20 in a stable fashion. These cells become less sensitive to inflammatory stimuli. This may be of interest in cell-based therapeutic approaches for clinical settings where inflammation is an important pathogenic factor.


Asunto(s)
Células Endoteliales/inmunología , Células Madre Fetales/inmunología , Inflamación/prevención & control , Proteínas Nucleares/biosíntesis , Transducción Genética , Proteínas Angiogénicas/genética , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Proteínas de Unión al ADN , Selectina E/genética , Selectina E/metabolismo , Sangre Fetal/citología , Sangre Fetal/inmunología , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Lentivirus/genética , Rodamiento de Leucocito , Monocitos/inmunología , Neovascularización Fisiológica/genética , Proteínas Nucleares/genética , ARN Mensajero/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
Histol Histopathol ; 25(2): 197-203, 2010 02.
Artículo en Inglés | MEDLINE | ID: mdl-20017106

RESUMEN

Human Papillomavirus (HPV)-associated esophageal carcinoma (EC) is a high incidence tumor worldwide. Dendritic cell (DC)-based tumor vaccine is considered an alternative therapy to treat EC. Here we developed a DC-based vaccine by transfecting cord blood CD34+ stem cell-derived DC with HPV18E7 gene, observed its biological characteristics and the antigen-specific T-cell cytotoxicity on EC cells induced by HPV18E7-DC in vitro. Our results showed that 1) HPV18E7 gene transfer did not change the typical morphology of mature DC, 2) the representative phenotypes of mature DC (CD80, CD86, and CD83) were highly expressed in HPV18E7- DC (81.6%, 80.5%, and 86.6%, respectively), 3) the expression level of 18E7 protein in HPV18E7-DC was 47.5%, and 4) the specific cytotoxicity against EC cells was significantly higher than that in controls (p<0.01). This study indicates the possibility of a DC-based immunotherapy in HPV-associated EC.


Asunto(s)
Vacunas contra el Cáncer , Carcinoma/inmunología , Proteínas de Unión al ADN/inmunología , Células Dendríticas/inmunología , Neoplasias Esofágicas/inmunología , Células Madre Fetales/inmunología , Activación de Linfocitos , Proteínas Oncogénicas Virales/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos CD/análisis , Antígenos CD34/análisis , Antígeno B7-1/análisis , Antígeno B7-2/análisis , Carcinoma/genética , Carcinoma/virología , Línea Celular Tumoral , Proliferación Celular , Forma de la Célula , Técnicas de Cocultivo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/virología , Sangre Fetal/citología , Sangre Fetal/inmunología , Citometría de Flujo , Humanos , Inmunoglobulinas/análisis , Inmunofenotipificación , Glicoproteínas de Membrana/análisis , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/genética , Transfección , Antígeno CD83
18.
Catheter Cardiovasc Interv ; 73(7): 917-24, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19455667

RESUMEN

Cellular cardiomyoplasty is undergoing intensive investigation as a new form of therapy for severely damaged hearts. Among several cell types, mesenchymal stem cells (MSCs) have been proposed as a potential cell source. MSC can be found in adult tissues or in fetal tissues like the umbilical chord blood, amniotic membrane, or amniotic fluid (AF). AF-MSCs have properties intermediate between embryonic and adult MSC, which make them particularly attractive for cellular regeneration. It has been shown that MSC could differentiate in cardiomyocytes-like cells in vitro. In some animal models, it has also been shown that transplanted MSC could engraft and show some cardiomyocytes-like characteristics. Since MSC do not express HLA-DR and present in vitro and in vivo immunosuppressive properties, they can be envisioned to be used in allogenic cellular cardiomyoplasty. Based on these promises, MSC from adult donors are currently used in small safety and feasibility trials. No clinical trial using AF-MSC has been performed yet. Still, the exact role of true cell repopulation and in situ cardiomyocytes differentiation versus pure paracrine effect after cell transplantation is currently much debated. Cellular cardiomyoplasty is a fascinating new area of investigation in regenerative medicine. Although considerable knowledge has been gained over the last decade on the use of MSC as a potential stem cell (SC) source, many issues remain unsolved. Because of several limitations in animal models, clinical studies in highly selected patients balancing the risks and benefits are required. In that regard, MSCs obtained from the fetal AF are a potential new source of SCs that need to be further investigated for cellular cardiomyoplasty.


Asunto(s)
Células Madre Adultas/trasplante , Líquido Amniótico/citología , Cardiomioplastia/métodos , Células Madre Fetales/trasplante , Cardiopatías/cirugía , Trasplante de Células Madre Mesenquimatosas , Miocitos Cardíacos/trasplante , Adulto , Células Madre Adultas/inmunología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Separación Celular , Células Madre Fetales/inmunología , Humanos , Miocitos Cardíacos/inmunología , Regeneración
19.
Circ Res ; 104(9): 1095-102, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19342601

RESUMEN

We evaluated the healing potential of human fetal aorta-derived CD133(+) progenitor cells and their conditioned medium (CD133(+) CCM) in a new model of ischemic diabetic ulcer. Streptozotocin-induced diabetic mice underwent bilateral limb ischemia and wounding. One wound was covered with collagen containing 2x10(4) CD133(+) or CD133(-) cells or vehicle. The contralateral wound, covered with only collagen, served as control. Fetal CD133(+) cells expressed high levels of wingless (Wnt) genes, which were downregulated following differentiation into CD133(-) cells along with upregulation of Wnt antagonists secreted frizzled-related protein (sFRP)-1, -3, and -4. CD133(+) cells accelerated wound closure as compared with CD133(-) or vehicle and promoted angiogenesis through stimulation of endothelial cell proliferation, migration, and survival by paracrine effects. CD133(+) cells secreted high levels of vascular endothelial growth factor (VEGF)-A and interleukin (IL)-8. Consistently, CD133(+) CCM accelerated wound closure and reparative angiogenesis, with this action abrogated by co-administering the Wnt antagonist sFRP-1 or neutralizing antibodies against VEGF-A or IL-8. In vitro, these effects were recapitulated following exposure of high-glucose-primed human umbilical vein endothelial cells to CD133(+) CCM, resulting in stimulation of migration, angiogenesis-like network formation and induction of Wnt expression. The promigratory and proangiogenic effect of CD133(+) CCM was blunted by sFRP-1, as well as antibodies against VEGF-A or IL-8. CD133(+) cells stimulate wound healing by paracrine mechanisms that activate Wnt signaling pathway in recipients. These preclinical findings open new perspectives for the cure of diabetic ulcers.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Pie Diabético/cirugía , Células Madre Fetales/trasplante , Isquemia/complicaciones , Extremidad Inferior/irrigación sanguínea , Neovascularización Fisiológica , Trasplante de Células Madre , Proteínas Wnt/metabolismo , Cicatrización de Heridas , Antígeno AC133 , Animales , Antígenos CD/análisis , Aorta/embriología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/cirugía , Pie Diabético/etiología , Pie Diabético/metabolismo , Pie Diabético/fisiopatología , Células Madre Fetales/inmunología , Células Madre Fetales/metabolismo , Glicoproteínas/análisis , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-8/metabolismo , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/cirugía , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Comunicación Paracrina , Péptidos/análisis , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Exp Clin Transplant ; 7(1): 50-5, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19364313

RESUMEN

OBJECTIVES: Cell populations residing in waste tissues (cord blood, umbilical cord, and placenta) may be collected without any medical or ethical contraindications concerning the mother or newborn baby. Cord blood hematopoietic stem cells are routinely used for clinical transplants; however, the low cell dose of the graft limits their therapeutic efficacy as it is associated with increased delayed or failed engraftment. The cell dose can be increased, and the efficacy of cord blood transplant potentially improved, by ex vivo expansion before transplant. MATERIALS AND METHODS: Twelve umbilical cord blood samples were included. The effect of cord blood storage at -80 degrees C on CD34+ cell count (mean -/+ standard deviation [SD]), cell viability (mean -/+ SD percent), and cell cycle status (percent quiescent versus dividing) was estimated. Ex vivo culture of cord blood mononuclear cells was done before storage, and after 1 week of freezing, and after 2 weeks of freezing. Ex vivo liquid culture was performed with media supplemented with stem cell factor, interleukin-3 (IL-3), and both. RESULTS: The count of CD34+ cells in pre-expansion aliquots decreased from 15.00 +/- 9.96 x 106 cells before freezing to 7.70 -/+ 3.20 x 106 cells after 2 weeks of freezing (P = .024). Cell viability in pre-expansion aliquots decreased from 99.5% -/+ 1.0% before freezing, to 52.5% -/+ 27.5% after 1 week of freezing (P = .013) and to 32.5% -/+ 9.5% after 2 weeks of freezing (P = .001). Mean fold of cell expansion and proportion of quiescent versus dividing cells did not change significantly from before to after freezing, and was not significantly different for culture with stem cell factor, IL-3, or both. CONCLUSION: Although freezing decreased cell count and viability, it did not impair the expansion potential of cord blood hematopoietic cells. Whether IL-3 or stem cell factor should be considered as essential components of expansion media is uncertain.


Asunto(s)
Proliferación Celular , Separación Celular , Criopreservación , Sangre Fetal/citología , Células Madre Fetales/fisiología , Células Madre Hematopoyéticas/fisiología , Antígenos CD34/análisis , Recuento de Células , Ciclo Celular , Supervivencia Celular , Células Cultivadas , Medios de Cultivo , Femenino , Células Madre Fetales/inmunología , Células Madre Fetales/metabolismo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-3/metabolismo , Embarazo , Factor de Células Madre/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...