Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 20699, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36450835

RESUMO

Kidney organoids derived from human induced pluripotent stem cells (iPSCs) have proven to be a valuable tool to study kidney development and disease. However, the lack of vascularization of these organoids often leads to insufficient oxygen and nutrient supply. Vascularization has previously been achieved by implantation into animal models, however, the vasculature arises largely from animal host tissue. Our aim is to transition from an in vivo implantation model towards an in vitro model that fulfils the advantages of vascularization whilst being fully human-cell derived. Our chip system supported culturing of kidney organoids, which presented nephron structures. We also showed that organoids cultured on chip showed increased maturation of endothelial populations based on a colocalization analysis of endothelial markers. Moreover, we observed migration and proliferation of human umbilical vein endothelial cells (HUVECs) cultured in the channels of the chip inside the organoid tissue, where these HUVECs interconnected with endogenous endothelial cells and formed structures presenting an open lumen resembling vessels. Our results establish for the first-time vascularization of kidney organoids in HUVEC co-culture conditions using a microfluidic organ-on-chip. Our model therefore provides a useful insight into kidney organoid vascularization in vitro and presents a tool for further studies of kidney development and drug testing, both for research purposes and pre-clinical applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Organoides , Animais , Humanos , Rim , Néfrons , Neovascularização Patológica , Células Endoteliais da Veia Umbilical Humana
2.
J Control Release ; 350: 642-651, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36063958

RESUMO

Vesicle-based medicines hold great promise for therapy development but essential knowledge on the bio-distribution and longevity of vesicles after administration is lacking. We generated vesicles from the membranes of human mesenchymal stromal cells (MSC) and we demonstrated earlier that these so-called membrane particles (MP) mediate immunomodulatory and regenerative responses in target cells. In the present study we examined the bio-distribution and longevity of MP after intravenous administration in mice. While most vesicle tracking methods are based on imaging techniques, which require labeling of vesicles and can only detect dense accumulations of vesicles, we used proteomics analysis to detect the presence of MP-derived proteins in multiple organs and tissues. MP proteins were mainly present in plasma and leukocytes at 1 h after injection, indicating that MP - in contrast to whole MSC - do not accumulate in the lungs upon first passage but remain in circulation. After 24 h, MP proteins were still present in plasma but were most abundant in the liver. RNA sequencing of livers demonstrated that MP impact liver function and in particular induce metabolic pathways. These data provide a clear view of the bio-distribution and longevity of MP, which is likely extrapolatable to other types of vesicles, and demonstrate that MP circulate for up to 24 h and may be a tool for targeting the liver.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Animais , Vesículas Extracelulares/metabolismo , Humanos , Imunomodulação , Fígado , Células-Tronco Mesenquimais/metabolismo , Camundongos
3.
Sci Rep ; 11(1): 8915, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33903694

RESUMO

The diagnosis of kidney allograft rejection is based on late histological and clinical markers. Early, specific and minimally-invasive biomarkers may improve rejection diagnosis. Endothelial cells (EC) are one of the earliest targets in kidney transplant rejection. We investigated whether circulating EC (cEC) could serve as an earlier and less invasive biomarker for allograft rejection. Blood was collected from a cohort of 51 kidney transplant recipients before and at multiple timepoints after transplantation, including during a for cause biopsy. The number and phenotype of EC was assessed by flow-cytometric analysis. Unbiased selection of EC was done using principal component (PCA) analysis. Paired analysis revealed a transient cEC increase of 2.1-fold on the third day post-transplant, recovering to preoperative levels at seventh day post-transplant and onwards. Analysis of HLA subtype demonstrated that cEC mainly originate from the recipient. cEC levels were not associated with allograft rejection, allograft function or other allograft pathologies. However, cEC in patients with allograft rejection and increased levels of cEC showed elevated levels of KIM-1 (kidney injury marker-1). These findings indicate that cEC numbers and phenotype are affected after kidney transplantation but may not improve rejection diagnosis.


Assuntos
Células Endoteliais/metabolismo , Citometria de Fluxo , Rejeição de Enxerto/sangue , Receptor Celular 1 do Vírus da Hepatite A/sangue , Transplante de Rim , Adulto , Aloenxertos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Tempo
4.
BJS Open ; 5(1)2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33609374

RESUMO

BACKGROUND: The 5-year graft survival rate of donor kidneys transplanted in the Eurotransplant Senior Program (ESP) is only 47 per cent. Normothermic machine perfusion (NMP) may be a new preservation technique that improves graft outcome. This pilot study aimed to assess safety and feasibility of this technique within the ESP. METHODS: Recipients were eligible for inclusion if they received a donor kidney within the ESP. Donor kidneys underwent 2 h of oxygenated NMP with a red cell-based solution at 37°C, additional to standard-of-care preservation (non-oxygenated hypothermic machine perfusion). The primary outcome was the safety and feasibility of NMP. As a secondary outcome, graft outcome was investigated and compared with that in a historical group of patients in the ESP and the contralateral kidneys. RESULTS: Eleven patients were included in the NMP group; the function of eight kidneys could be compared with that of the contralateral kidney. Fifty-three patients in the ESP, transplanted consecutively between 2016 and 2018, were included as controls. No adverse events were noted, especially no arterial thrombosis or primary non-function of the transplants. After 120 min of oxygenated NMP, median flow increased from 117 (i.q.r. 80-126) to 215 (170-276) ml/min (P = 0.001). The incidence of immediate function was 64 per cent in the NMP group and 40 per cent in historical controls (P = 0.144). A significant difference in graft outcome was not observed. DISCUSSION: This pilot study showed NMP to be safe and feasible in kidneys transplanted in the ESP. A well powered study is warranted to confirm these results and investigate the potential advantages of NMP on graft outcome.


Assuntos
Transplante de Rim/métodos , Rim , Preservação de Órgãos/métodos , Perfusão/métodos , Idoso , Função Retardada do Enxerto , Estudos de Viabilidade , Feminino , Rejeição de Enxerto , Sobrevivência de Enxerto , Humanos , Transplante de Rim/efeitos adversos , Masculino , Preservação de Órgãos/instrumentação , Perfusão/instrumentação , Projetos Piloto , Estudos Prospectivos
5.
Osteoarthritis Cartilage ; 27(12): 1831-1840, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31536814

RESUMO

OBJECTIVE: Synovium contains multipotent progenitor/stromal cells (MPCs) with potential to participate in cartilage repair. Understanding the identity of these MPCs will allow their therapeutic potential to be fully exploited. Hence this study aimed to identify primary synovial MPCs and characterize them in the context of cartilage regeneration. METHODS: Primary MPC/MPC-subset specific markers in synovium were identified by FACS analysis of uncultured cells. MPC-subsets from human synovium obtained from patients undergoing total knee arthroplasty were FACS sorted, cultured, immunophenotyped and chondrogenically differentiated. The anatomical localization of MPCs in synovium was examined using immunohistochemistry. Finally, the presence of these MPC subsets in healthy synovium obtained from human organ donors was examined. RESULTS: A combination of CD45, CD31, CD73 and CD90 can isolate two distinct MPC-subsets in synovium. These MPC-subsets, freshly isolated from synovium, did not express CD45 or CD31, but expressed CD73. Additionally, a sub-population of CD73+ cells also expressed CD90. CD45-CD31-CD73+CD90- cells were significantly more chondrogenic than CD45-CD31-CD73+CD90+ cells in the presence of TGFß1. Interestingly, reduced chondrogenic ability of CD73+CD90+ cells could be reversed by the addition of BMP2, showing discrete chondrogenic factor requirements by distinct cell-subsets. In addition, these MPCs had distinct anatomical localization; CD73 was expressed both in intimal and sub-intimal region while CD90 was enriched in the sub-intimal region. We further demonstrated that these subsets are also present in healthy synovium. CONCLUSIONS: We provide indications that primary MPCs in synovial intima and sub-intima are phenotypically and functionally distinct with different chondrogenic properties.


Assuntos
Cartilagem Articular/fisiologia , Diferenciação Celular/fisiologia , Condrogênese/fisiologia , Células-Tronco Multipotentes/metabolismo , Osteoartrite do Joelho , Regeneração/fisiologia , 5'-Nucleotidase/metabolismo , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Moléculas de Adesão Celular/metabolismo , Feminino , Citometria de Fluxo , Proteínas Ligadas por GPI/metabolismo , Humanos , Imuno-Histoquímica , Imunofenotipagem , Antígenos Comuns de Leucócito/metabolismo , Masculino , Pessoa de Meia-Idade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Membrana Sinovial/citologia , Antígenos Thy-1/metabolismo
6.
Eur Cell Mater ; 33: 43-58, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-28138954

RESUMO

Bone marrow derived mesenchymal stem cells (MSCs) have immunomodulatory and trophic capacities. For therapeutic application in local chronic inflammatory diseases, MSCs, preferably of allogeneic origin, have to retain immunomodulatory properties. This might be achieved by encapsulation of MSCs in a biomaterial that protects them from the host immune system. Most studies investigating the properties of MSCs for therapeutic application use short term cultures of cells in monolayer. Since the physical environment of MSCs can influence their functionality, we evaluated the feasibility of preserving the immunomodulatory properties of MSCs encapsulated in a three-dimensional alginate construct. After 5 weeks of implantation in immunocompetent rats, active allogeneic MSCs encapsulated in alginate were still detectable by Bio Luminescence Imaging and Magnetic Resonance Imaging of luciferase transduced and superparamagnetic iron oxide labelled MSCs. MSCs injected in saline were only detectable up to 1 week after injection. Moreover, the MSCs encapsulated in alginate responded to inflammatory stimuli similarly to MSCs in monolayer culture. In addition, MSC-alginate beads secreted immunomodulatory and trophic factors and inhibited T-cell proliferation after 30 d of in vitro culture. Our data indicate that allogeneic MSCs encapsulated in alginate persist locally and could act as an interactive immunomodulatory or trophic factor release system for several weeks, making this an interesting system to investigate for application in inflammatory disease conditions.


Assuntos
Alginatos/farmacologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Adipogenia/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/metabolismo , Meios de Cultivo Condicionados/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/farmacologia , Humanos , Imunocompetência/efeitos dos fármacos , Imunomodulação/efeitos dos fármacos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Ratos Wistar , Tela Subcutânea/efeitos dos fármacos , Tela Subcutânea/patologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Transplante Homólogo
7.
Stem Cells Int ; 2016: 5761487, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26981132

RESUMO

Liver ischemia reperfusion injury (IRI) is inevitable during transplantation and resection and is characterized by hepatocellular injury. Therapeutic strategies to reduce IRI and accelerate regeneration could offer major benefits. Mesenchymal stem cells (MSC) are reported to have anti-inflammatory and regeneration promoting properties. We investigated the effect of MSC in a model of combined IRI and partial resection in the mouse. Hepatic IRI was induced by occlusion of 70% of the blood flow during 60 minutes, followed by 30% hepatectomy. 2 × 10(5) MSC or PBS were infused 2 hours before or 1 hour after IRI. Six, 48, and 120 hours postoperatively mice were sacrificed. Liver damage was evaluated by liver enzymes, histology, and inflammatory markers. Regeneration was determined by liver/body weight ratio, proliferating hepatocytes, and TGF-ß levels. Fate of MSC was visualized with 3D cryoimaging. Infusion of 2 × 10(5) MSC 2 hours before or 1 hour after IRI and resection showed no beneficial effects. Tracking revealed that MSC were trapped in the lungs and did not migrate to the site of injury and many cells had already disappeared 2 hours after infusion. Based on these findings we conclude that intravenously infused MSC disappear rapidly and were unable to induce beneficial effects in a clinically relevant model of IRI and resection.

8.
Clin Exp Immunol ; 179(3): 509-19, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25310899

RESUMO

Renal tubular epithelial cells (TECs) are one of the main targets of alloreactive T cells during acute rejection. We hypothesize that TECs modulate the outcome of alloimmunity by executing immunosuppressive effects in order to dampen the local inflammation. We studied whether TECs possess immunosuppressive capacities and if indoleamine 2,3-dioxygenase (IDO) might play a role in suppressing T cell alloreactivity. Next, we studied the role of programmed death ligand 1 (PD-L1) and intercellular adhesion molecule-1 (ICAM-1 with regard to TEC-related immunomodulatory effects. CD3/CD28 and alloactivated peripheral blood mononuclear cells were co-cultured with activated TECs. We analysed CD4(+) and CD8(+) T cell proliferation and apoptosis in the absence or presence of IDO inhibitor 1-methyl-L-tryptophan (1-L-MT), anti-PD-L1 and anti-ICAM-1. Further, we examined whether inhibition of T cell proliferation was cell-cell contact-dependent. We found that TECs dose-dependently inhibited CD4(+) and CD8(+) T cell proliferation (P<0.05). Activated TECs showed significantly increased IDO activity and up-regulated PD-L1 and ICAM-1 expression. Suppressed CD4(+) and CD8(+) T cell proliferation was only partially restored or failed to restore using 1-L-MT. Activated TECs increased early and late apoptosis of proliferating CD4(+) and CD8(+) T cells; only CD4(+) T cell apoptosis was statistically affected by 1-L-MT. Transwell experiments revealed that TEC-mediated immunosuppression is cell-cell contact-dependent. We found that anti-ICAM-1 affected only CD4(+) T cell apoptosis and not T cell proliferation. Our data show that TECs suppress both CD4(+) and CD8(+) T cell proliferation contact dependently. Interestingly, inhibition of proliferation and enhancement of apoptosis of T cell subsets is differentially regulated by indoleamine 2,3-dioxygenase and ICAM-1, with no evidence for the involvement of PD-L1 in our system.


Assuntos
Antígeno B7-H1/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Epiteliais/imunologia , Rejeição de Enxerto/tratamento farmacológico , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Transplante de Rim , Túbulos Renais/patologia , Anticorpos Bloqueadores/farmacologia , Apoptose/efeitos dos fármacos , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Comunicação Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Ativação Enzimática/efeitos dos fármacos , Adesões Focais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Rejeição de Enxerto/imunologia , Humanos , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Isoantígenos/imunologia , Triptofano/análogos & derivados , Triptofano/farmacologia
9.
Stem Cells ; 33(3): 880-91, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25376628

RESUMO

Mesenchymal or stromal stem cells (MSC) interact with cells of the immune system in multiple ways. Modulation of the immune system by MSC is believed to be a therapeutic option for autoimmune disease and transplant rejection. In recent years, B cells have moved into the focus of the attention as targets for the treatment of immune disorders. Current B-cell targeting treatment is based on the indiscriminate depletion of B cells. The aim of this study was to examine whether human adipose tissue-derived MSC (ASC) interact with B cells to affect their proliferation, differentiation, and immune function. ASC supported the survival of quiescent B cells predominantly via contact-dependent mechanisms. Coculture of B cells with activated T helper cells led to proliferation and differentiation of B cells into CD19(+) CD27(high) CD38(high) antibody-producing plasmablasts. ASC inhibited the proliferation of B cells and this effect was dependent on the presence of T cells. In contrast, ASC directly targeted B-cell differentiation, independently of T cells. In the presence of ASC, plasmablast formation was reduced and IL-10-producing CD19(+) CD24(high) CD38(high) B cells, known as regulatory B cells, were induced. These results demonstrate that ASC affect B cell biology in vitro, suggesting that they can be a tool for the modulation of the B-cell response in immune disease.


Assuntos
Tecido Adiposo/citologia , Linfócitos B Reguladores/citologia , Comunicação Celular/imunologia , Células-Tronco Mesenquimais/citologia , Plasmócitos/citologia , Linfócitos T Auxiliares-Indutores/citologia , Tecido Adiposo/imunologia , Apoptose/imunologia , Linfócitos B Reguladores/imunologia , Diferenciação Celular/imunologia , Processos de Crescimento Celular/imunologia , Sobrevivência Celular/imunologia , Técnicas de Cocultura , Humanos , Células-Tronco Mesenquimais/imunologia , Tonsila Palatina/citologia , Tonsila Palatina/imunologia , Plasmócitos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia
10.
Clin Exp Immunol ; 174(3): 449-58, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24028656

RESUMO

CD28/B7 co-stimulation blockade with belatacept prevents alloreactivity in kidney transplant patients. However, cells lacking CD28 are not susceptible to belatacept treatment. As CD8(+) CD28(-) T-cells have cytotoxic and pathogenic properties, we investigated whether mesenchymal stem cells (MSC) are effective in controlling these cells. In mixed lymphocyte reactions (MLR), MSC and belatacept inhibited peripheral blood mononuclear cell (PBMC) proliferation in a dose-dependent manner. MSC at MSC/effector cell ratios of 1:160 and 1:2·5 reduced proliferation by 38·8 and 92·2%, respectively. Belatacept concentrations of 0·1 µg/ml and 10 µg/ml suppressed proliferation by 20·7 and 80·6%, respectively. Both treatments in combination did not inhibit each other's function. Allostimulated CD8(+) CD28(-) T cells were able to proliferate and expressed the cytolytic and cytotoxic effector molecules granzyme B, interferon (IFN)-γ and tumour necrosis factor (TNF)-α. While belatacept did not affect the proliferation of CD8(+) CD28(-) T cells, MSC reduced the percentage of CD28(-) T cells in the proliferating CD8(+) T cell fraction by 45·9% (P = 0·009). CD8(+) CD28(-) T cells as effector cells in MLR in the presence of CD4(+) T cell help gained CD28 expression, an effect independent of MSC. In contrast, allostimulated CD28(+) T cells did not lose CD28 expression in MLR-MSC co-culture, suggesting that MSC control pre-existing CD28(-) T cells and not newly induced CD28(-) T cells. In conclusion, alloreactive CD8(+) CD28(-) T cells that remain unaffected by belatacept treatment are inhibited by MSC. This study indicates the potential of an MSC-belatacept combination therapy to control alloreactivity.


Assuntos
Antígenos CD28/metabolismo , Linfócitos T CD8-Positivos/imunologia , Imunoconjugados/farmacologia , Leucócitos Mononucleares/imunologia , Células-Tronco Mesenquimais/imunologia , Abatacepte , Apoptose , Proliferação de Células , Células Cultivadas , Granzimas/biossíntese , Humanos , Interferon gama/biossíntese , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos , Fator de Necrose Tumoral alfa/biossíntese
11.
Clin Exp Immunol ; 173(2): 343-54, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23607314

RESUMO

Due to their immunomodulatory properties, mesenchymal stem cells (MSC) are interesting candidates for cellular therapy for autoimmune disorders, graft-versus-host disease and allograft rejection. MSC inhibit the proliferation of effector T cells and induce T cells with a regulatory phenotype. So far it is unknown whether human MSC-induced CD4(+) CD25(+) CD127(-) forkhead box P3 (FoxP3)(+) T cells are functional and whether they originate from effector T cells or represent expanded natural regulatory T cells (nT(reg)). Perirenal adipose-tissue derived MSC (ASC) obtained from kidney donors induced a 2·1-fold increase in the percentage of CD25(+) CD127(-) FoxP3(+) cells within the CD4(+) T cell population from allostimulated CD25(-/dim) cells. Interleukin (IL)-2 receptor blocking prevented this induction. The ASC-induced T cells (iT(reg)) inhibited effector cell proliferation as effectively as nT(reg). The vast majority of cells within the iT(reg) fraction had a methylated FOXP3 gene T(reg)-specific demethylated region (TSDR) indicating that they were not of nT(reg) origin. In conclusion, ASC induce T(reg) from effector T cells. These iT(reg) have immunosuppressive capacities comparable to those of nT(reg). Their induction is IL-2 pathway-dependent. The dual effect of MSC of inhibiting immune cell proliferation while generating de-novo immunosuppressive cells emphasizes their potential as cellular immunotherapeutic agent.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Mesenquimais/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Tecido Adiposo/citologia , Anticorpos Monoclonais/farmacologia , Basiliximab , Antígenos CD4/metabolismo , Separação Celular , Células Cultivadas , Metilação de DNA , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Humanos , Tolerância Imunológica , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Receptores de Interleucina-2/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia
12.
Front Immunol ; 3: 297, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056000

RESUMO

Mesenchymal stem cells (MSC) are under investigation as a therapy for a variety of disorders. Although animal models show long term regenerative and immunomodulatory effects of MSC, the fate of MSC after infusion remains to be elucidated. In the present study the localization and viability of MSC was examined by isolation and re-culture of intravenously infused MSC. C57BL/6 MSC (500,000) constitutively expressing DsRed-fluorescent protein and radioactively labeled with Cr-51 were infused via the tail vein in wild-type C57BL/6 mice. After 5 min, 1, 24, or 72 h, mice were sacrificed and blood, lungs, liver, spleen, kidneys, and bone marrow removed. One hour after MSC infusion the majority of Cr-51 was found in the lungs, whereas after 24 h Cr-51 was mainly found in the liver. Tissue cultures demonstrated that viable donor MSC were present in the lungs up to 24 h after infusion, after which they disappeared. No viable MSC were found in the other organs examined at any time. The induction of ischemia-reperfusion injury in the liver did not trigger the migration of viable MSC to the liver. These results demonstrate that MSC are short-lived after i.v. infusion and that viable MSC do not pass the lungs. Cell debris may be transported to the liver. Long term immunomodulatory and regenerative effects of infused MSC must therefore be mediated via other cell types.

13.
Front Immunol ; 3: 212, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22833744

RESUMO

The research on T cell immunosuppression therapies has attracted most of the attention in clinical transplantation. However, B cells and humoral immune responses are increasingly acknowledged as crucial mediators of chronic allograft rejection. Indeed, humoral immune responses can lead to renal allograft rejection even in patients whose cell-mediated immune responses are well controlled. On the other hand, newly studied B cell subsets with regulatory effects have been linked to tolerance achievement in transplantation. Better understanding of the regulatory and effector B cell responses may therefore lead to new therapeutic approaches. Mesenchymal stem cells (MSC) are arising as a potent therapeutic tool in transplantation due to their regenerative and immunomodulatory properties. The research on MSCs has mainly focused on their effects on T cells and although data regarding the modulatory effects of MSCs on alloantigen-specific humoral response in humans is scarce, it has been demonstrated that MSCs significantly affect B cell functioning. In the present review we will analyze and discuss the results in this field.

14.
Hum Gene Ther ; 22(12): 1587-91, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21732766

RESUMO

Mesenchymal stem cells (MSCs) have potential for therapeutic application as an immunomodulatory and regenerative agent. The immunogenicity and survival of MSCs after infusion are, however, not clear and evidence suggests that allogeneic but also autologous MSCs disappear rapidly after infusion. This may be associated with the susceptibility of MSCs to lysis by natural killer (NK) cells, possibly a result of culture-induced stress. In the present study we examined whether NK cell-mediated lysis of MSCs could be inhibited by immunosuppressive drugs. Human MSCs were isolated from adipose tissue and expanded in culture. Peripheral blood mononuclear cells were activated with interleukin (IL)-2 (200 U/ml) and IL-15 (10 ng/ml) for 7 days. CD3(-)CD16(+)CD56(+) NK cells were then isolated by fluorescence-activated cell sorting and added to europium-labeled MSCs for 4 hr in the presence or absence of immunosuppressive drugs. Lysis of MSCs was determined by spectrophotometric measurement of europium release. Nonactivated NK cells were not capable of lysing MSCs. Cytokine-activated NK cells showed upregulated levels of granzyme B and perforin and efficiently lysed allogeneic and autologous MSCs. Addition of tacrolimus, rapamycin or sotrastaurin to the lysis assay did not inhibit MSC killing. Furthermore, preincubation of activated NK cells with the immunosuppressive drugs for 24 hr before exposure to MSCs had no effect on MSC lysis. Last, addition of the immunosuppressants before and during the activation of NK cells, reduced NK cell numbers but did not affect their capacity to lyse MSCs. We conclude that the immunosuppressive drugs tacrolimus, rapamycin, and sotrastaurin are not capable of inhibiting the lysis of allogeneic and autologous MSCs by activated NK cells. Other approaches to controlling lysis of MSCs should be investigated, as controlling lysis may determine the efficacy of MSC therapy.


Assuntos
Citotoxicidade Imunológica/imunologia , Células Matadoras Naturais/imunologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/farmacologia , Citometria de Fluxo , Humanos , Imunossupressores/farmacologia , Interleucina-15/farmacologia , Interleucina-2/farmacologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Células-Tronco Mesenquimais/efeitos dos fármacos , Sirolimo/farmacologia , Tacrolimo/farmacologia , Transplante Autólogo , Transplante Homólogo
15.
Clin Exp Immunol ; 162(3): 474-86, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20846162

RESUMO

There is emerging interest in the application of mesenchymal stem cells (MSC) for the prevention and treatment of autoimmune diseases, graft-versus-host disease and allograft rejection. It is, however, unknown how inflammatory conditions affect phenotype and function of MSC. Adipose tissue-derived mesenchymal stem cells (ASC) were cultured with alloactivated peripheral blood mononuclear cells (PBMC) (mixed lymphocyte reaction: MLR), with proinflammatory cytokines [interferon (IFN)-γ, tumour necrosis factor (TNF)-α and interleukin (IL)-6] or under control conditions, and their full genome expression and function examined. Proinflammatory cytokines mainly increased indoleamine-2,3-dioxygenase expression, whereas ASC cultured with MLR showed increased expression of COX-2, involved in prostaglandin E(2) production. Both conditions had a stimulatory, but differential, effect on the expression of proinflammatory cytokines and chemokines, while the expression of fibrotic factors was decreased only in response to proinflammatory cytokines. Functional analysis demonstrated that inflammatory conditions affected morphology and proliferation of ASC, while their differentiation capacity and production of trophic factors was unaffected. The immunosuppressive capacity of ASC was enhanced strongly under inflammatory conditions. In conclusion, ASC showed enhanced immunosuppressive capacity under inflammatory conditions, while their differentiation capacity was preserved. Therefore, in vitro preconditioning provides ASC with improved properties for immediate clinical immune therapy.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Citocinas/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Mediadores da Inflamação/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Tecido Adiposo/citologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Ciclo-Oxigenase 2/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Humanos , Terapia de Imunossupressão , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo
16.
Am J Transplant ; 9(1): 222-30, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18976299

RESUMO

Mesenchymal stem cells (MSC) are characterized by their multilineage differentiation capacity and immunosuppressive properties. They are resident in virtually all tissues and we have recently characterized MSC from the human heart. Clinical heart transplantation offers a model to study the fate of transplanted human MSC. In this study, we isolated and expanded MSC from heart tissue taken before, and 1 week up to 6 years after heart transplantation. MSC from posttransplantation tissue were all of donor origin, demonstrating the longevity of endogenous MSC and suggesting an absence of immigration of recipient MSC into the heart. MSC isolated from transplanted tissue showed an immunophenotype that was characteristic for MSC and maintained cardiomyogenic and osteogenic differentiation capacity. They furthermore preserved their ability to inhibit the proliferative response of donor-stimulated recipient peripheral blood mononuclear cells. In conclusion, functional MSC of donor origin remain present in the heart for several years after transplantation.


Assuntos
Transplante de Coração , Células-Tronco Mesenquimais/citologia , Miocárdio/patologia , Doadores de Tecidos , Diferenciação Celular , Linhagem da Célula , Citometria de Fluxo , Genótipo , Humanos , Imunofenotipagem , Teste de Cultura Mista de Linfócitos , Células-Tronco Mesenquimais/imunologia
17.
Stem Cells Dev ; 16(4): 597-604, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17784833

RESUMO

Mesenchymal stem cells (MSCs) have important tissue repair functions and show potent immunosuppressive capacities in vitro. Although usually isolated from the bone marrow, MSCs have been identified in other tissues, including the skin and liver. In the present study, we isolated and characterized MSCs from human heart, spleen, and perirenal adipose tissue. MSCs from these different tissue sites were similar to those derived from bone marrow in that they expressed comparable levels of the cell-surface markers CD90, CD105, CD166, and HLA class I, were negative for CD34, CD45, HLA class II, CD80, and CD86 expression, and were capable of osteogenic and adipogenic differentiation. Like bone marrow-derived MSCs, MSCs from these different tissue sources inhibited the proliferation of alloactivated peripheral blood mononuclear cells (PBMCs), giving 85%, 79%, 79%, and 81% inhibition, respectively. Also in line with bone marrow-derived MSCs they inhibited proliferative responses of PBMCs to phytohemagglutinin, a nonspecific stimulator of lymphocyte proliferation, and reduced-memory T lymphocyte responses to tetanus toxoid. The results of this study demonstrate that MSCs from various tissues have similar immunophenotypes, in vitro immunosuppressive properties, and differentiation potential.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Células-Tronco Mesenquimais/imunologia , Miocárdio/citologia , Miocárdio/imunologia , Baço/citologia , Baço/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Técnicas de Cultura de Células , Divisão Celular , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/fisiologia , Citometria de Fluxo , Humanos , Leucócitos Mononucleares/citologia , Fígado/citologia , Fígado/imunologia , Teste de Cultura Mista de Linfócitos , Células-Tronco Mesenquimais/citologia , Pele/citologia , Pele/imunologia
18.
Pigment Cell Res ; 19(1): 58-67, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16420247

RESUMO

Glutamate is the major excitatory neurotransmitter in the central nervous system but has also important functions in the epidermis. It is involved in keratinocyte barrier function and in re-epithelialization processes after wounding. Recently, glutamate signalling has been suggested to be implicated in the development of melanoma. The present study examined the expression and functionality of metabotropic and ionotropic glutamate receptors on normal human melanocytes. We found that cultured melanocytes expressed the ionotropic glutamate receptors GluR2 and 4 [alpha-amino-3-hydroxy-5-methyl-4-isoxsazolepropionic acid (AMPA) receptors] and N-methyl-d-aspartate (NMDA) receptors 2A and 2C and possibly the metabotropic glutamate receptor 1. Melanocytes were also found to express specific glutamate transporters and decarboxylases, but appeared neither to produce nor to release l-glutamate. Stimulation with 10 or 100 microM AMPA or NMDA elevated intracellular calcium concentrations in melanocytes, and thus demonstrated the functionality of the glutamate receptors. Millimolar concentrations of l-glutamate did not induce melanocyte toxicity and had no stimulating effect on melanin production. However, blockage of AMPA and NMDA receptors with CFM-2, memantine or MK801 caused a rapid and reversible change in melanocyte morphology, which was associated with disorganisation of actin and tubulin microfilaments. After 24 h of treatment with the AMPA receptor inhibitor CFM-2, there was a sharp reduction in the expression of the crucial melanocyte differentiation and proliferation factor MiTF. The results of this study demonstrate a role for glutamate in melanocyte regulation that may have implications in melanocyte associated disorders.


Assuntos
Regulação da Expressão Gênica , Melanócitos/metabolismo , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Receptores de Glutamato/metabolismo , Apoptose/fisiologia , Benzodiazepinonas/farmacologia , Forma Celular , Células Cultivadas , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Perfilação da Expressão Gênica , Ácido Glutâmico/metabolismo , Humanos , Melanócitos/citologia , Melanócitos/efeitos dos fármacos , Memantina/farmacologia , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Glutamato/genética
19.
Exp Cell Res ; 294(1): 60-7, 2004 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-14980501

RESUMO

Reactive oxygen species (ROS) such as hydrogen peroxide (H(2)O(2)) are produced in the skin under the influence of UV radiation. These compounds are highly reactive and can induce DNA lesions in epidermal cells. Melanin is considered to protect human skin against DNA damage by absorbing UV radiation. We have investigated whether melanin can, in addition, offer protection against the effects of H(2)O(2) in human melanocytes and HaCaT keratinocytes. In the present study, it was shown that 40 and 100 microM H(2)O(2) increased the number of DNA strand breaks as measured using the comet assay, in melanocytes of Caucasian origin. In melanocytes of the same origin in which melanin levels were increased by culturing in presence of 10 mM NH(4)Cl and elevated l-tyrosine, H(2)O(2)-induced DNA damage was reduced compared to that in control melanocytes. Similarly, HaCaT cells that were loaded with melanin were better protected against H(2)O(2)-induced DNA strand breaks than control HaCaT cells. These protective effects of melanin were mimicked by the intracellular Ca(2+)-chelator BAPTA. Thus, BAPTA reduced the level of H(2)O(2)-induced DNA strand breaks in melanocytes. Like BAPTA, melanin is known to be a potent chelator of Ca(2+) and this was confirmed in the present study. It was shown that melanin levels in melanocytic cells correlated directly with intracellular Ca(2+) binding capacity and, in addition, correlated inversely with H(2)O(2)-induced increases in intracellular Ca(2+). Our results show that melanin may have an important role in regulating intracellular Ca(2+) homeostasis and it is suggested that melanin protects against H(2)O(2)-induced DNA strand breaks in both melanocytes and keratinocytes and through its ability to bind Ca(2+).


Assuntos
Cálcio/metabolismo , Dano ao DNA , Ácido Egtázico/análogos & derivados , Peróxido de Hidrogênio/toxicidade , Queratinócitos/metabolismo , Melaninas/fisiologia , Melanócitos/metabolismo , Linhagem Celular , Células Cultivadas , Quelantes/farmacologia , Ensaio Cometa , Citoproteção , Ácido Egtázico/farmacologia , Humanos , Peróxido de Hidrogênio/antagonistas & inibidores , Queratinócitos/efeitos dos fármacos , Queratinócitos/ultraestrutura , Melaninas/metabolismo , Melanócitos/efeitos dos fármacos , Melanócitos/ultraestrutura
20.
Pigment Cell Res ; 16(2): 127-32, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12622789

RESUMO

We have examined whether melanin affects Ca2+ homeostasis in cultured normal human melanocytes. Intracellular Ca2+ concentrations ([Ca2+]i), were measured in four Caucasian and in three Negroid melanocyte cultures. Under resting conditions [Ca2+]i was around 100 nM in all cultures, but differences between cells within cultures were observed. All cultures responded to endothelin-1 (ET-1) with increases in [Ca2+]i and there were no differences between Caucasian and Negroid cultures. However, large differences in responses between cells within cultures were observed, indicating that melanocyte cultures are very heterogeneous. The addition of 2.5 mM CaCl2 to melanocytes kept in Ca2+-free medium resulted in rapid and transient increases in [Ca2+]i of up to 1500 nM. These increases were on average more than two times smaller in melanocyte cultures established from Negroid donors compared with Caucasian cultures. In addition, well melanized Caucasian melanocytes, cultured in the presence of 400 microM tyrosine and 10 mM NH4Cl, showed a reduced increase in cytoplasmic Ca2+ concentration upon the addition of extracellular Ca2+. The difference in maintaining Ca2+ homeostasis between poorly and well melanized melanocytes may be the result of the clearance of cytoplasmic Ca2+ into melanosomes and the greater capacity for this in the more pigmented melanocytes.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Epiderme/metabolismo , Homeostase/fisiologia , Melaninas/metabolismo , Melanócitos/metabolismo , Cloreto de Amônio/farmacologia , População Negra/genética , Cloreto de Cálcio/farmacologia , Células Cultivadas , Endotelina-1/farmacologia , Células Epidérmicas , Humanos , Líquido Intracelular/metabolismo , Masculino , Melanócitos/citologia , Melanócitos/efeitos dos fármacos , Tirosina/farmacologia , População Branca/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...