Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biomed Res Int ; 2013: 151679, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24369528

RESUMO

To evaluate the potential therapeutic effect of the infusion of hMSCs for the correction of liver injuries, we performed total body radiation exposure of NOD/SCID mice. After irradiation, mir-27b level decreases in liver, increasing the directional migration of hMSCs by upregulating SDF1 α . A significant increase in plasmatic transaminases levels, apoptosis process in the liver vascular system, and in oxidative stress were observed. hMSC injection induced a decrease in transaminases levels and oxidative stress, a disappearance of apoptotic cells, and an increase in Nrf2, SOD gene expression, which might reduce ROS production in the injured liver. Engrafted hMSCs expressed cytokeratin CK18 and CK19 and AFP genes indicating possible hepatocyte differentiation. The presence of hMSCs expressing VEGF and Ang-1 in the perivascular region, associated with an increased expression of VEGFr1, r2 in the liver, can confer a role of secreting cells to hMSCs in order to maintain the endothelial function. To explain the benefits to the liver of hMSC engraftment, we find that hMSCs secreted NGF, HGF, and anti-inflammatory molecules IL-10, IL1-RA contributing to prevention of apoptosis, increasing cell proliferation in the liver which might correct liver dysfunction. MSCs are potent candidates to repair and protect healthy tissues against radiation damages.


Assuntos
Diferenciação Celular , Hepatócitos/citologia , Fígado/lesões , Células-Tronco Mesenquimais/citologia , Animais , Antioxidantes/metabolismo , Quimiocina CXCL12/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Fígado/efeitos da radiação , Células-Tronco Mesenquimais/metabolismo , Camundongos , MicroRNAs/metabolismo , Irradiação Corporal Total
2.
Methods Mol Biol ; 826: 179-88, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22167649

RESUMO

This work was initiated in an effort to evaluate the potential therapeutic contribution of the infusion of mesenchymal stem cells (MSC) for the correction of liver injuries. We subjected NOD-SCID mice to a 10.5-Gy abdominal irradiation and we tested the biological and histological markers of liver injury in the absence and after infusion of expanded human MSC. Irradiation alone induced a significant elevation of the ALT and AST. Apoptosis in the endothelial layer of vessels was observed. When MSC were infused in mice, a significant decrease of transaminases was measured, and a total disappearance of apoptotic cells. MSC were not found in liver. To explain the protection of liver without MSC engraftment, we hypothesize an indirect action of MSC on the liver via the intestinal tract. Pelvic or total body irradiation induces intestinal absorption defects leading to an alteration of the enterohepatic recirculation of bile acids. This alteration induces an increase in Deoxy Cholic Acid (DCA) which is hepatoxic. In this study, we confirm these results. DCA concentration increased approximately twofold after irradiation but stayed to the baseline level after MSC injection. We propose from our observations that, following irradiation, MSC infusion indirectly corrected liver dysfunction by preventing gut damage. This explanation would be consistent with the absence of MSC engraftment in liver. These results evidenced that MSC treatment of a target organ may have an effect on distant tissues. This observation comes in support to the interest for the use of MSC for cellular therapy in multiple pathologies proposed in the recent years.


Assuntos
Fígado/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Lesões Experimentais por Radiação/terapia , Alanina Transaminase/sangue , Análise de Variância , Animais , Aspartato Aminotransferases/sangue , Ácidos e Sais Biliares/análise , Citometria de Fluxo , Técnicas Histológicas , Humanos , Marcação In Situ das Extremidades Cortadas , Fígado/efeitos da radiação , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Lesões Experimentais por Radiação/patologia , Reação em Cadeia da Polimerase em Tempo Real
3.
Stem Cells Dev ; 17(6): 1165-74, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19006457

RESUMO

The aim of this work was to characterize multipotent mesenchymal stromal cells (MSCs) in the postnatal human thymus and to localize these MSCs in the organ. Adherent cells isolated from thymus samples were characterized by cell-surface antigen expression. This showed that adherent cells have a MSC profile as assessed by the expression of CD73 and CD105 markers and the lack of CD45 expression. These cells are able to differentiate in vitro into adipocytes, osteoblasts, and chondrocytes and to inhibit mixed lymphocyte reaction. This indicates that isolated cells have all of the characteristics of MSC. The fibroblast colony-forming unit (CFU-F) assay was used to determine their frequency in the postnatal thymus. This frequency was 60.9 +/- 14.8 CFU-F per 1 x 10(5) freshly isolated mononuclear cells. Moreover, taking advantage of CD34 and CD105 expression, immunohistological staining allowed us to localize MSC within interlobular trabeculae in close contact with the outer cortex. Polymerase chain reaction experiments indicated that thymic MSC expressed interleukin-7 and stromal cell-derived factor-1 messenger RNA. Overall, these results confirm previous findings of the presence in the adult human thymus of multipotent MSCs with a phenotype similar to adipose-derived adult stem cells. These results also show for the first time a histological localization of MSC in an organ. This suggests a possible role of thymic MSC in intrathymic differentiation.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Timo/citologia , Timo/metabolismo , 5'-Nucleotidase/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Condrócitos/citologia , Condrócitos/metabolismo , Ensaio de Unidades Formadoras de Colônias/métodos , Endoglina , Feminino , Humanos , Lactente , Recém-Nascido , Interleucina-7/biossíntese , Antígenos Comuns de Leucócito/metabolismo , Linfocinas/biossíntese , Masculino , Osteoblastos/citologia , Osteoblastos/metabolismo , Fator de Crescimento Derivado de Plaquetas/biossíntese , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Células Estromais/citologia , Células Estromais/metabolismo
4.
Int J Radiat Biol ; 84(8): 659-67, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18608642

RESUMO

PURPOSE: To define the ability of human bone marrow (BM) stromal cells to produce fms-like tyrosine kinase 3 (Flt3)-ligand (FL), and the effect of irradiation, tumour necrosis factor-alpha (TNFalpha) or tumour growth factor beta (TGFbeta) on FL production. MATERIAL AND METHODS: Primary BM stromal cell cultures were irradiated at 2-10 Gy or were stimulated with TNFalpha or TGFbeta1. The presence of FL was tested in culture supernatants and in cell lysate. The presence of a membrane-bound form of FL and the level of gene expression were also tested. RESULTS: Primary BM stromal cells spontaneously released FL. This production was increased by TNFalpha but not by TGFbeta1 or by irradiation. Chemical induction of osteoblastic differentiation from BM stromal cells also induced an increase in FL release. CONCLUSIONS: Our results suggest that the observed increase in FL concentration after in vivo irradiation is an indirect effect. The possible implication of BM stromal cells in these mechanisms is discussed.


Assuntos
Células da Medula Óssea/metabolismo , Proteínas de Membrana/biossíntese , Fator de Crescimento Transformador beta1/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos da radiação , Diferenciação Celular , Células Cultivadas , Humanos , Osteoblastos/citologia , Fenótipo , Células Estromais/metabolismo , Células Estromais/efeitos da radiação
5.
Ann Hematol ; 86(1): 1-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17043780

RESUMO

It has been suggested that human mesenchymal stem cells (hMSC) could be used to repair numerous injured tissues. We have studied the potential use of hMSC to limit radiation-induced skin lesions. Immunodeficient NOD/SCID mice were locally irradiated to the leg (30 Gy, dose rate 2.7 Gy/min) using a (60)Co source to induce a severe skin lesion. Cultured bone marrow hMSC were delivered intravenously to the mice. The irradiated skin samples were studied for the presence of the human cells, the severity of the lesions and the healing process. Macroscopic analysis and histology results showed that the lesions were evolving to a less severe degree of radiation dermatitis after hMSC transplant when compared to irradiated non-transplanted controls. Clinical scores for the studied skin parameters of treated mice were significantly improved. A faster healing was observed when compared to untreated mouse. Immunohistology and polymerase chain reaction analysis provided evidence that the human cells were found in the irradiated area. These results suggest a possible use of hMSC for the treatment of the early phase of the cutaneous radiation syndrome. A successful transplant of stem cells and subsequent reduction in radiation-induced complication may open the road to completely new strategies in cutaneous radiation syndrome therapy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Radiodermite/terapia , Transplante Heterólogo , Animais , Células da Medula Óssea/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Radiodermite/patologia , Síndrome , Cicatrização
6.
Adv Exp Med Biol ; 585: 19-30, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17120774

RESUMO

Patients who undergo pelvic or abdominal radiotherapy may develop side effects that can be life threatening. Tissue complications caused by radiation-induced stem cell depletion may result in structural and functional alterations of the gastrointestinal (GI) tract. Stem cell therapy using mesenchymal stem cells (MSC) is a promising approach for replenishment of the depleted stem cell compartment during radiotherapy. There is little information on the therapeutic potential of MSC in injured-GI tract following radiation exposure. In this study, we addressed the ability of MSC to support the structural regeneration of the small intestine after abdominal irradiation. We isolated MSC from human bone marrow and human mesenchymal stem cells (hMSC) were transplanted into immunotolerent NOD/SCID mice with a dose of 5.10(6) cells via the systemic route. Using a model of radiation-induced intestinal injury, we studied the link between damage, hMSC engraftment and the capacity of hMSC to sustain structural recovery. Tissue injury was assessed by histological analysis. hMSC engraftment in tissues was quantified by PCR assay. Following abdominal irradiation, the histological analysis of small intestinal structure confirms the presence of partial and transient (three days) mucosal atrophy. PCR analysis evidences a low but significant hMSC implantation in small intestine (0.17%) but also at all the sites of local irradiation (kidney, stomach and spleen). Finally, in presence of hMSC, the small intestinal structure is already recovered at three days after abdominal radiation exposure. We show a structural recovery accompanied by an increase of small intestinal villus height, three and fifteen days following abdominal radiation exposure. In this study, we show that radiation-induced small intestinal injury may play a role in the recruitment of MSC for the improvement of tissue recovery. This work supports, the use of MSC infusion to repair damaged GI tract in patients subjected to radiotherapy. MSC therapy to avoid extended intestinal crypt sterilization is a promising approach to diminish healthy tissue alterations during the course of pelvic radiotherapy.


Assuntos
Células Epiteliais/citologia , Intestino Delgado/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Lesões Experimentais por Radiação , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Células Epiteliais/efeitos da radiação , Humanos , Intestino Delgado/patologia , Intestino Delgado/efeitos da radiação , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos da radiação , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Regeneração , Transplante de Células-Tronco/métodos
7.
Stem Cells ; 24(4): 1020-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16339642

RESUMO

Mesenchymal stem cells (MSCs) have been shown to migrate to various tissues. There is little information on the fate and potential therapeutic efficacy of the reinfusion of MSCs following total body irradiation (TBI). We addressed this question using human MSC (hMSCs) infused to nonobese diabetic/ severe combined immunodeficient (NOD/SCID) mice submitted to TBI. Further, we tested the impact of additional local irradiation (ALI) superimposed to TBI, as a model of accidental irradiation. NOD/SCID mice were transplanted with hM-SCs. Group 1 was not irradiated before receiving hMSC infusion. Group 2 received only TBI at a dose of 3.5 Gy, group 3 received local irradiation to the abdomen at a dose of 4.5 Gy in addition to TBI, and group 4 received local irradiation to the leg at 26.5 Gy in addition to TBI. Fifteen days after irradiation, quantitative and spatial distribution of the hMSCs were studied. Histological analysis of mouse tissues confirmed the presence of radio-induced lesions in the irradiated fields. Following their infusion into nonirradiated animals, hMSCs homed at a very low level to various tissues (lung, bone marrow, and muscles) and no significant engraftment was found in other organs. TBI induced an increase of engraftment levels of hMSCs in the brain, heart, bone marrow, and muscles. Abdominal irradiation (AI) as compared with leg irradiation (LI) increased hMSC engraftment in the exposed area (the gut, liver, and spleen). Hind LI as compared with AI increased hMSC engraftment in the exposed area (skin, quadriceps, and muscles). An increase of hMSC engraftment in organs outside the fields of the ALI was also observed. Conversely, following LI, hMSC engraftment was increased in the brain as compared with AI. This study shows that engraftment of hMSCs in NOD/ SCID mice with significantly increased in response to tissue injuries following TBI with or without ALI. ALI induced an increase of the level of engraftment at sites outside the local irradiation field, thus suggesting a distant (abscopal) effect of radiation damage. This work supports the use of MSCs to repair damaged normal tissues following accidental irradiation and possibly in patients submitted to radiotherapy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos da radiação , Animais , Movimento Celular/efeitos da radiação , Expressão Gênica , Globinas/genética , Sobrevivência de Enxerto/genética , Sobrevivência de Enxerto/efeitos da radiação , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Especificidade de Órgãos , Lesões Experimentais por Radiação/patologia , Lesões Experimentais por Radiação/terapia , Transplante Heterólogo , Irradiação Corporal Total , Microglobulina beta-2/metabolismo
8.
Genet Vaccines Ther ; 2: 16, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15509303

RESUMO

BACKGROUND: Targeted gene transfection remains a crucial issue to permit the real development of genetic therapy. As such, in vivo targeted transfection of specific subsets of hematopoietic stem cells might help to sustain hematopoietic recovery from bone marrow aplasia by providing local production of growth factors. METHODS: Balb/C mice were injected intravenously, with an anti-mouse c-kit (CD117) monoclonal antibody chemically coupled to a human IL-3 gene-containing plasmid DNA. Mice were sacrificed for tissue analyses at various days after injection of the conjugates. RESULTS: By ELISA, the production of human IL-3 was evidenced in the sera of animals 5 days after treatment. Cytofluorometric analysis after in vivo transfection of a reporter gene eGFP demonstrated transfection of CD117+/Sca1+ hematopoietic immature cells. By PCR analysis of genomic DNA and RNA using primer specific pIL3 sequences, presence and expression of the human IL-3-transgene were detected in the bone marrow up to 10 days in transfected mice but not in control animals. CONCLUSIONS: These data clearly indicate that antibody-mediated endocytosis gene transfer allows the expression of the IL-3 transgene into hematopoietic immature cells, in vivo. While availability of marketed recombinant growth factors is restricted, this targeting strategy should permit delivery of therapeutic genes to tissues of interest through systemic delivery. In particular, the ability to specifically target growth factor expression into repopulating hematopoietic stem cells may create new opportunities for the treatment of primary or radiation-induced marrow failures.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA