Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Geriatr Oncol ; 14(7): 101567, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37453811

RESUMO

INTRODUCTION: Blood biomarkers are potentially useful prognostic markers and may support treatment decisions, but it is unknown if and which biomarkers are most useful in older patients with solid tumors. The aim of this systematic review was to evaluate the evidence on the association of blood biomarkers with treatment response and adverse health outcomes in older patients with solid tumors. MATERIALS AND METHODS: A literature search was conducted in five databases in December 2022 to identify studies on blood biomarkers measured before treatment initiation, not tumor specific, and outcomes in patients with solid tumors aged ≥60 years. Studies on any type or line of oncologic treatment could be included. Titles and abstracts were screened by three authors. Data extraction and quality assessment, using the Quality in Prognosis Studies (QUIPS) checklist, were performed by two authors. RESULTS: Sixty-three studies were included, with a median sample size of 138 patients (Interquartile range [IQR] 99-244) aged 76 years (IQR 72-78). Most studies were retrospective cohort studies (63%). The risk of bias was moderate in 52% and high in 43%. Less than one-third reported geriatric parameters. Eighty-six percent examined mortality outcomes, 37% therapeutic response, and 37% adverse events. In total, 77 unique markers were studied in patients with a large variety of tumor types and treatment modalities. Neutrophil-to-lymphocyte ratio (20 studies), albumin (19), C-reactive protein (16), hemoglobin (14) and (modified) Glasgow Prognostic Score ((m)GPS) (12) were studied most often. The vast majority showed no significant association of these biomarkers with outcomes, except for associations between low albumin and adverse events and high (m)GPS with mortality. DISCUSSION: Most studies did not find a significant association between blood biomarkers and clinical outcomes. The interpretation of current evidence on prognostic blood biomarkers is hampered by small sample sizes and inconsistent results across heterogeneous studies. The choice for blood biomarkers in the majority of included studies seemed driven by availability in clinical practice in retrospective cohort studies. Ageing biomarkers are rarely studied in older patients with solid tumors. Further research is needed in larger and more homogenous cohorts that combine clinical parameters and biomarkers before these can be used in clinical practice.


Assuntos
Neoplasias , Humanos , Idoso , Estudos Retrospectivos , Neoplasias/terapia , Prognóstico , Biomarcadores , Avaliação de Resultados em Cuidados de Saúde
2.
Int J Mol Sci ; 20(14)2019 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-31340593

RESUMO

Normothermic machine perfusion (NMP) of kidneys offers the opportunity to perform active interventions, such as the addition of mesenchymal stromal cells (MSCs), to an isolated organ prior to transplantation. The purpose of this study was to determine whether administering MSCs to kidneys during NMP is feasible, what the effect of NMP is on MSCs and whether intact MSCs are retained in the kidney and to which structures they home. Viable porcine kidneys were obtained from a slaughterhouse. Kidneys were machine perfused during 7 h at 37 °C. After 1 h of perfusion either 0, 105, 106 or 107 human adipose tissue derived MSCs were added. Additional ex vivo perfusions were conducted with fluorescent pre-labelled bone-marrow derived MSCs to assess localisation and survival of MSCs during NMP. After NMP, intact MSCs were detected by immunohistochemistry in the lumen of glomerular capillaries, but only in the 107 MSC group. The experiments with fluorescent pre-labelled MSCs showed that only a minority of glomeruli were positive for infused MSCs and most of these glomeruli contained multiple MSCs. Flow cytometry showed that the number of infused MSCs in the perfusion circuit steeply declined during NMP to approximately 10%. In conclusion, the number of circulating MSCs in the perfusate decreases rapidly in time and after NMP only a small portion of the MSCs are intact and these appear to be clustered in a minority of glomeruli.


Assuntos
Rastreamento de Células/métodos , Glomérulos Renais/ultraestrutura , Transplante de Rim , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Perfusão/métodos , Adipócitos/citologia , Adipócitos/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Diferenciação Celular , Corantes Fluorescentes/metabolismo , Humanos , Glomérulos Renais/cirurgia , Transplante de Células-Tronco Mesenquimais/instrumentação , Células-Tronco Mesenquimais/fisiologia , Microscopia de Fluorescência , Preservação de Órgãos/métodos , Compostos Orgânicos/metabolismo , Perfusão/instrumentação , Suínos , Temperatura , Transplante Heterólogo
3.
Stem Cells Dev ; 25(18): 1342-54, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27349989

RESUMO

Mesenchymal stem cells (MSC) are studied as a cell therapeutic agent for treatment of various immune diseases. However, therapy with living culture-expanded cells comes with safety concerns. Furthermore, development of effective MSC immunotherapy is hampered by lack of knowledge of the mechanisms of action and the therapeutic components of MSC. Such knowledge allows better identification of diseases that are responsive to MSC treatment, optimization of the MSC product, and development of therapy based on functional components of MSC. To close in on the components that carry the therapeutic immunomodulatory activity of MSC, we generated MSC that were unable to respond to inflammatory signals or secrete immunomodulatory factors, but preserved their cellular integrity [heat-inactivated MSC (HI-MSC)]. Secretome-deficient HI-MSC and control MSC showed the same biodistribution and persistence after infusion in mice with ischemic kidney injury. Both control and HI-MSC induced mild inflammatory responses in healthy mice and dramatic increases in interleukin-10, and reductions in interferon gamma levels in sepsis mice. In vitro experiments showed that opposite to control MSC, HI-MSC lacked the capability to suppress T-cell proliferation or induce regulatory B-cell formation. However, both HI-MSC and control MSC modulated monocyte function in response to lipopolysaccharides. The results of this study demonstrate that, in particular disease models, the immunomodulatory effect of MSC does not depend on their secretome or active cross-talk with immune cells, but on recognition of MSC by monocytic cells. These findings provide a new view on MSC-induced immunomodulation and help identify key components of the therapeutic effects of MSC.


Assuntos
Imunomodulação , Células-Tronco Mesenquimais/imunologia , Animais , Linfócitos B/citologia , Movimento Celular , Proliferação de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imunofenotipagem , Inflamação/patologia , Infusões Intravenosas , Lipopolissacarídeos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Monócitos/citologia , Sepse/imunologia , Sepse/patologia , Linfócitos T/citologia , Distribuição Tecidual
4.
Stem Cells Dev ; 23(19): 2328-35, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25105211

RESUMO

Mesenchymal stem cells (MSC) are present in the bone marrow, from where they are thought to migrate through the blood stream to the sites of injury. However, virtually all tissues contain resident MSC that may contribute to local regenerative and immunomodulatory processes, thereby hypothetically preempting the need for recruiting MSC through the bloodstream. Although there is some indication for circulating MSC in animal models, there is little solid evidence for the mobilization and migration of MSC in the human circulation. In the present study, we were unable to detect MSC in the blood of healthy individuals. We then searched for MSC in the blood of ten patients with end-stage renal disease, ten patients with end-stage liver disease, and in eight heart transplant patients with biopsy-proven rejection by culturing of mononuclear cells under MSC-supporting culture conditions. In none of these patient categories, MSC were identified in the blood. MSC were, however, found in the blood of a severe trauma patient with multiple fractures, suggesting that disruption of bone marrow leads to the release of MSC into the blood stream. The conclusion of this study is that MSC are not recruited into the circulation in patients with injured solid organs and during aggressive immune responses after transplantation.


Assuntos
Movimento Celular , Fraturas Ósseas/patologia , Células-Tronco Mesenquimais/citologia , Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Fraturas Ósseas/metabolismo , Humanos , Leucócitos/citologia
5.
Cytotherapy ; 15(11): 1352-61, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24094487

RESUMO

BACKGROUND AIMS: Adipose tissue-derived mesenchymal stromal cells (ASCs) are of interest as a cell therapeutic agent for immunologic and degenerative diseases. During in vitro expansion, ASCs may be at risk for genetic alterations, and genetic screening is a prerequisite. We examined the presence of aneuploidy in ASCs and its origin and development during culture and evaluated the implications of aneuploidy for therapeutic use of ASCs. METHODS: Adipose tissue of healthy individuals was used for isolation and expansion of ASCs. Chromosome copy numbers were studied using fluorescence in situ hybridization analysis. Aneuploidy was studied in freshly isolated ASCs, in ASCs cultured for 0-16 passages and in senescent cultures. To evaluate the plasticity of ploidy, ASCs were cloned, and the variation of ploidy in the clones was examined. Tumorigenicity was studied by subcutaneous injection of aneuploid ASCs in immunodeficient NOD/SCID mice. RESULTS: No aneuploidy was detected in freshly isolated ASCs. In low passages (passages 0-4), aneuploidy was detected in 3.4% of ASCs. Prolonged culture expansion of ASCs (passages 5-16) resulted in a significant increase of aneuploidy to 7.1%. With senescence, aneuploidy increased further to 19.8%. Aneuploidy was observed in clones of diploid ASCs, demonstrating the de novo development of aneuploidy. No transformation of ASCs was observed, and in contrast to cancer cell lines, aneuploid ASCs were incapable of tumor formation in immunodeficient mice. CONCLUSIONS: ASC cultures contain a stable percentage of aneuploid cells. Aneuploidy was not a predecessor of transformation or tumor formation. This finding indicates that aneuploidy is culture-induced but unlikely to compromise clinical application of ASCs.


Assuntos
Tecido Adiposo/citologia , Aneuploidia , Transformação Celular Neoplásica/genética , Instabilidade Genômica/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Adulto , Idoso , Animais , Diferenciação Celular , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Senescência Celular , Células Clonais/citologia , Feminino , Humanos , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade
6.
Front Immunol ; 4: 203, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23882269

RESUMO

Adipose tissue-derived mesenchymal stem cells (ASC) are of great interest as a cellular therapeutic agent for regenerative and immunomodulatory purposes. The function of ASC adapts to environmental conditions, such as oxygen tension. Oxygen levels within tissues are typically much lower than under standard culture conditions and ASC used for therapy therefore encounter a change from normoxic to hypoxic conditions. The effect of hypoxia on the regenerative potential of ASC has been investigated in a number of studies. The effect of hypoxia on the immunomodulatory function of ASC, however, remains to be determined. In the present study the effect of hypoxic (1% oxygen) culture conditions on human ASC was examined. ASC showed no signs of toxicity under low oxygen levels and no major immunophenotypical changes were observed, apart from a down regulation of the marker CD105. Oxygen tension had no effect on the proliferation of ASC and colony forming unit efficiency remained the same under 1 and 20% oxygen. Under both oxygen levels ASC were capable of strong upregulation of the immunomodulatory molecules indoleamine 2,3-dioxygenase (IDO) and programed death ligand-1 upon stimulation with IFN-γ and TNF-α, and, in addition, IDO activity as measured by the accumulation of l-kynurenine was not affected under hypoxia. The ability of ASC to inhibit anti-CD3/CD28 stimulated CD4(+) and CD8(+) T cell proliferation was not hampered by hypoxia. The results of the present study demonstrate that the immunosuppressive capacity of ASC is maintained under hypoxic conditions. These findings are important for the therapeutic use of ASC and may be applied for the in vitro generation of ASC with improved functionality for therapeutic use.

7.
Transplantation ; 96(3): 234-8, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23759879

RESUMO

The Fourth Expert Meeting of the Mesenchymal Stem Cells in Solid Organ Transplantation (MiSOT) Consortium took place in Barcelona on October 19 and 20, 2012. This meeting focused on the translation of preclinical data into early clinical settings. This position paper highlights the main topics explored on the safety and efficacy of mesenchymal stem cells as a therapeutic agent in solid organ transplantation and emphasizes the issues (proper timing, concomitant immunossupression, source and immunogenicity of mesenchymal stem cells, and oncogenicity) that have been addressed and will be followed up by the MiSOT Consortium in future studies.


Assuntos
Transplante de Células-Tronco Mesenquimais , Ensaios Clínicos como Assunto , Humanos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/legislação & jurisprudência
8.
Stem Cells Dev ; 22(21): 2825-35, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23767885

RESUMO

Mesenchymal stem cells (MSCs) have potent immunosuppressive effects in vitro and are considered as a therapeutic option for autoimmune disease and organ transplantation. While MSCs show beneficial effects on immune disease progression and transplant survival in animal models, the immunomodulatory mechanisms involved are largely unknown. In the present study, we show that intravenously infused C57BL/6- green fluorescent protein (GFP) MSCs home to the lungs in C57BL/6 recipient mice and induce an inflammatory response. This response was characterized by increased mRNA expression of monocyte chemoattractant protein-1 (MCP1), IL1-ß, and TNF-α and an increase in macrophages in lung tissue 2 h after MSC infusion. Simultaneously, serum levels of proinflammatory IL6, CXCL1, and MCP1 protein increased, demonstrating systemic immune activation after MSC infusion. In liver tissue, no C57BL/6-GFP MSCs were detected, but MCP1 and TNF-α mRNA levels peaked 4 h after MSC infusion. The expression of the anti-inflammatory cytokines TGF-ß, IL4, and IL10 was only marginally affected. Nevertheless, 3 days after MSC infusion, animals developed a milder inflammatory response to lipopolysaccharides. Our results suggest that the in vivo immunomodulatory effects of MSCs originate from an inflammatory response that is induced by the infusion of MSCs, which is followed by a phase of reduced immune reactivity.


Assuntos
Quimiocinas/imunologia , Citocinas/imunologia , Pulmão/imunologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/imunologia , Tecido Adiposo/citologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiocinas/sangue , Quimiocinas/genética , Citocinas/sangue , Citocinas/genética , Expressão Gênica/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Mediadores da Inflamação/sangue , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Infusões Intravenosas , Rim/imunologia , Rim/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Fígado/imunologia , Fígado/metabolismo , Pulmão/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Fatores de Tempo
9.
Transpl Int ; 26(6): 651-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23682671

RESUMO

Mesenchymal stem cells (MSCs) possess immunomodulatory properties which are of key interest for their application in autoimmunity and transplantation. In transplantation, administration of MSCs has shown promising results in preclinical models and has recently moved to clinical trials. Therefore, it is important to study the interactions between MSCs and immunosuppressive drugs currently used in transplantation. We aimed to analyze the effect of rabbit antithymocyte globulin (rATG) MSCs. MSCs were obtained from perirenal fat of kidney donors and exposed to ranging doses of rATG (Thymoglobulin(®) , Genzyme; 0.5-100 µg/ml). Binding of rATG, effects on viability and susceptibility to be killed by cytotoxic lymphocytes as well as effects on their immunosuppressive potential of MSCs were tested. rATG binds dose-dependently to MSCs. This binding was associated with slightly impaired viability after 48 and 72 h when compared with nonexposed MSCs. In contrast to nontreated MSCs, rATG preexposed MSCs were susceptible to be lysed by cytokine-activated CD8(+) cytotoxic cells and NKT cells. The capacity of MSCs to suppress the proliferation of anti-CD3/CD28 activated CD4 and CD8 T cells were reduced by the presence of rATG in the culture. rATG reduces the viability and antiproliferative capacity of MSCs in a dose-dependent manner and converts them into targets for CD8 T cells and NKT cell lysis.


Assuntos
Soro Antilinfocitário/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Soro Antilinfocitário/metabolismo , Linfócitos T CD8-Positivos/imunologia , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imunossupressores/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Coelhos
10.
Cytotherapy ; 15(6): 663-72, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23419679

RESUMO

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) are pluripotent cells that have immunosuppressive and reparative properties in vitro and in vivo. Although autologous bone marrow (BM)-derived MSCs are already clinically tested in transplant recipients, it is unclear whether these BM cells are affected by renal disease. We assessed whether renal failure affected the function and therapeutic potential of BM-MSCs. METHODS: MSCs from 10 adults with end-stage renal disease (ESRD) and 10 age-matched healthy controls were expanded from BM aspirates and tested for phenotype and functionality in vitro. RESULTS: MSCs from ESRD patients were >90% positive for CD73, CD90 and CD105 and negative for CD34 and CD45 and showed a similar morphology and differentiation capacity as MSCs from healthy controls. Of importance for their clinical utility, growth characteristics were similar in both groups, and sufficient numbers of MSCs were obtained within 4 weeks. Messenger RNA expression levels of self-renewal genes and factors involved in repair and inflammation were also comparable between both groups. Likewise, microRNA expression profiling showed a broad overlap between ESRD and healthy donor MSCs. ESRD MSCs displayed the same immunosuppressive capacities as healthy control MSCs, demonstrated by a similar dose-dependent inhibition of peripheral blood mononuclear cell proliferation, similar inhibition of proinflammatory cytokines tumor necrosis factor-α and interferon-γ production and a concomitant increase in the production of interleukin-10. CONCLUSIONS: Expanded BM-MSCs procured from ESRD patients and healthy controls are both phenotypically and functionally similar. These findings are important for the potential autologous clinical application of BM-MSCs in transplant recipients.


Assuntos
Células da Medula Óssea/citologia , Terapia Baseada em Transplante de Células e Tecidos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Idoso , Feminino , Humanos , Falência Renal Crônica/patologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Transplante Autólogo
11.
J Stem Cell Res Ther ; Suppl 6(1): 20780, 2013 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-24672744

RESUMO

BACKGROUND: Recent studies with bone marrow (BM)-derived Mesenchymal Stromal Cells (MSC) in transplant recipients demonstrate that treatment with MSC is safe and clinically feasible. While BM is currently the preferred source of MSC, adipose tissue is emerging as an alternative. To develop efficient therapies, there is a need for preclinical efficacy studies in transplantation. We used a unique humanized transplantation model to study the in vivo immunosuppressive effect of human BM-MSC and adipose tissue-derived MSC (ASC). METHODS: Gene expression of BM-MSC and ASC and their capacity to inhibit activated PBMC proliferation was evaluated. The in vivo immunosuppressive effect of BM-MSC and ASC was studied in a humanized mouse model. SCID mice were transplanted with human skin grafts and injected with human allogeneic PBMC with or without administration of BM-MSC or ASC. The effect of MSC on skin graft rejection was studied by immunohistochemistry and PCR. RESULTS: BM-MSC and ASC expressed TGFß, CXCL-10 and IDO. IDO expression and acitivity increased significantly in BM-MSC and ASC upon IFN-γ stimulation. IFN-γ stimulated BM-MSC and ASC inhibited the proliferation of activated PBMC in a significant and dose dependent manner. In our humanized mouse model, alloreactivity was marked by pronounced CD45+ T-cell infiltrates consisting of CD4+ and CD8+ T cells and increased IFN-γ expression in the skin grafts which were all significantly inhibited by both BM-MSC and ASC. CONCLUSION: BM-MSC and ASC are immunosuppressive in vitro and suppress alloreactivity in a preclinical humanized transplantation model.

12.
J Stem Cell Res Ther ; 3(Suppl 6): 004, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24729944

RESUMO

INTRODUCTION: For clinical applications, Mesenchymal Stromal Cells (MSC) can be isolated from bone marrow and adipose tissue of autologous or allogeneic origin. Allogeneic cell usage has advantages but may harbor the risk of sensitization against foreign HLA. Therefore, we evaluated whether bone marrow and adipose tissue-derived MSC are capable of inducing HLA-specific alloreactivity. METHODS: MSC were isolated from healthy human Bone Marrow (BM-MSC) and adipose tissue (ASC) donors. Peripheral Blood Mononuclear Cells (PBMC) were co-cultured with HLA-AB mismatched BM-MSC or ASC precultured with or without IFNy. After isolation via FACS sorting, the educated CD8+ T effector populations were exposed for 4 hours to Europium labeled MSC of the same HLA make up as in the co-cultures or with different HLA. Lysis of MSC was determined by spectrophotometric measurement of Europium release. RESULTS: CD8+ T cells educated with BM-MSC were capable of HLA specific lysis of BM-MSC. The maximum lysis was 24% in an effector:target (E:T) ratio of 40:1. Exposure to IFNγ increased HLA-I expression on BM-MSC and increased lysis to 48%. Co-culturing of PBMC with IFNγ-stimulated BM-MSC further increased lysis to 76%. Surprisingly, lysis induced by ASC was significantly lower. CD8+ T cells educated with ASC induced a maximum lysis of 13% and CD8+ T cells educated with IFNγ-stimulated ASC of only 31%. CONCLUSION: Allogeneic BM-MSC, and to a lesser extend ASC, are capable of inducing HLA specific reactivity. These results should be taken into consideration when using allogeneic MSC for clinical therapy.

13.
Kidney Int ; 82(7): 748-58, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22695328

RESUMO

Mesenchymal stem cells are a potential therapeutic agent in renal disease and kidney transplantation. Autologous cell use in kidney transplantation is preferred to avoid anti-HLA reactivity; however, the influence of renal disease on mesenchymal stem cells is unknown. To investigate the feasibility of autologous cell therapy in patients with renal disease, we isolated these cells from subcutaneous adipose tissue of healthy controls and patients with renal disease and compared them phenotypically and functionally. The mesenchymal stem cells from both groups showed similar morphology and differentiation capacity, and were both over 90% positive for CD73, CD105, and CD166, and negative for CD31 and CD45. They demonstrated comparable population doubling times, rates of apoptosis, and were both capable of inhibiting allo-antigen- and anti-CD3/CD28-activated peripheral blood mononuclear cell proliferation. In response to immune activation they both increased the expression of pro-inflammatory and anti-inflammatory factors. These mesenchymal stem cells were genetically stable after extensive expansion and, importantly, were not affected by uremic serum. Thus, mesenchymal stem cells of patients with renal disease have similar characteristics and functionality as those from healthy controls. Hence, our results indicate the feasibility of their use in autologous cell therapy in patients with renal disease.


Assuntos
Tecido Adiposo/metabolismo , Nefropatias/metabolismo , Células-Tronco Mesenquimais/metabolismo , 5'-Nucleotidase/metabolismo , Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Adulto , Idoso , Antígenos CD/metabolismo , Apoptose , Biomarcadores/metabolismo , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Estudos de Casos e Controles , Moléculas de Adesão Celular Neuronais/metabolismo , Diferenciação Celular , Proliferação de Células , Separação Celular/métodos , Forma Celular , Células Cultivadas , Técnicas de Cocultura , Endoglina , Feminino , Proteínas Fetais/metabolismo , Citometria de Fluxo , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica , Humanos , Imunofenotipagem/métodos , Mediadores da Inflamação/metabolismo , Nefropatias/sangue , Nefropatias/imunologia , Nefropatias/patologia , Antígenos Comuns de Leucócito/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Pessoa de Meia-Idade , Fenótipo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptores de Superfície Celular/metabolismo , Fatores de Tempo , Transplante Autólogo , Uremia/sangue , Uremia/imunologia
14.
Curr Opin Organ Transplant ; 17(1): 55-62, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22186092

RESUMO

PURPOSE OF REVIEW: Mesenchymal stem or stromal cells (MSCs) are multipotent cells with immunomodulatory and regenerative capacities. These properties make MSCs interesting for use as a cell therapeutic agent in solid-organ transplantation (SOT). Their applicability in SOT has recently been evaluated in numerous preclinical and clinical studies, which are reviewed in this article. RECENT FINDINGS: Animal transplant studies indicate that MSCs can improve the outcome of SOT. In-vitro research has resulted in new insights into the paracrine mechanism and cell-dependent mechanism of action, migratory behaviour and immunogenicity of MSCs that are relevant for application of MSCs in SOT. Currently, six trials are registered to evaluate MSCs in phase I/II studies in SOT. The first clinical data are now published describing MSC infusion in organ transplant recipients. SUMMARY: The research to date confirms that MSCs are a promising cell therapeutic agent for application in SOT and the first clinical trials have been initiated. The exact mechanisms of MSC therapy have, however, not been elucidated. Further basic and clinical research is needed to develop effective MSC therapy in SOT patients.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Transplante de Órgãos , Animais , Movimento Celular , Sobrevivência Celular , Ensaios Clínicos como Assunto , Humanos , Terapia de Imunossupressão/efeitos adversos , Imunossupressores/farmacologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA