Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cytotherapy ; 24(8): 774-788, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35613962

RESUMO

The ISCT Scientific Signature Series Symposium "Advances in Cell and Gene Therapies for Lung Diseases and Critical Illnesses" was held as an independent symposium in conjunction with the biennial meeting, "Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases," which took place July 12-15, 2021, at the University of Vermont. This is the third Respiratory System-based Signature Series event; the first 2, "Tracheal Bioengineering, the Next Steps" and "Cellular Therapies for Pulmonary Diseases and Critical Illnesses: State of the Art of European Science," took place in 2014 and 2015, respectively. Cell- and gene-based therapies for respiratory diseases and critical illnesses continue to be a source of great promise and opportunity. This reflects ongoing advancements in understanding of the mechanisms by which cell-based therapies, particularly those using mesenchymal stromal cells (MSCs), can mitigate different lung injuries and the increasing sophistication with which preclinical data is translated into clinical investigations. This also reflects continuing evolution in gene transfer vectors, including those designed for in situ gene editing in parallel with those targeting gene or cell replacement. Therefore, this symposium convened global thought leaders in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value of cell- and gene-based therapies for patients with respiratory diseases and critical illnesses.


Assuntos
Estado Terminal , Pneumopatias , Terapia Baseada em Transplante de Células e Tecidos , Estado Terminal/terapia , Terapia Genética , Humanos , Pneumopatias/genética , Pneumopatias/terapia , Células-Tronco
2.
Front Immunol ; 12: 716606, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539651

RESUMO

Recent clinical experience has demonstrated that adoptive regulatory T (Treg) cell therapy is a safe and feasible strategy to suppress immunopathology via induction of host tolerance to allo- and autoantigens. However, clinical trials continue to be compromised due to an inability to manufacture a sufficient Treg cell dose. Multipotent adult progenitor cells (MAPCⓇ) promote Treg cell differentiation in vitro, suggesting they may be repurposed to enhance ex vivo expansion of Tregs for adoptive cellular therapy. Here, we use a Good Manufacturing Practice (GMP) compatible Treg expansion platform to demonstrate that MAPC cell-co-cultured Tregs (MulTreg) exhibit a log-fold increase in yield across two independent cohorts, reducing time to target dose by an average of 30%. Enhanced expansion is coupled to a distinct Treg cell-intrinsic transcriptional program characterized by elevated expression of replication-related genes (CDK1, PLK1, CDC20), downregulation of progenitor and lymph node-homing molecules (LEF1 CCR7, SELL) and induction of intestinal and inflammatory tissue migratory markers (ITGA4, CXCR1) consistent with expression of a gut homing (CCR7lo ß7hi) phenotype. Importantly, we find that MulTreg are more readily expanded from patients with autoimmune disease compared to matched Treg lines, suggesting clinical utility in gut and/or T helper type1 (Th1)-driven pathology associated with autoimmunity or transplantation. Relative to expanded Tregs, MulTreg retain equivalent and robust purity, FoxP3 Treg-Specific Demethylated Region (TSDR) demethylation, nominal effector cytokine production and potent suppression of Th1-driven antigen specific and polyclonal responses in vitro and xeno Graft vs Host Disease (xGvHD) in vivo. These data support the use of MAPC cell co-culture in adoptive Treg therapy platforms as a means to rescue expansion failure and reduce the time required to manufacture a stable, potently suppressive product.


Assuntos
Autoimunidade , Contagem de Linfócitos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/imunologia , Células-Tronco Adultas/metabolismo , Animais , Doenças Autoimunes/etiologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Biomarcadores , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Regulação da Expressão Gênica , Doença Enxerto-Hospedeiro/diagnóstico , Doença Enxerto-Hospedeiro/etiologia , Humanos , Imunofenotipagem , Masculino , Camundongos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
3.
Stem Cells Transl Med ; 10(11): 1561-1574, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34397170

RESUMO

Cell-based therapy for the treatment of inflammatory disorders has focused on the application of mesenchymal stromal cells (MSCs) and multipotent adult progenitor cells (MAPCs). Despite the recent positive findings in industry-sponsored clinical trials of MSCs and MAPCs for graft vs host disease (GvHD), cell therapy is efficacious in some but not all patients, highlighting the need to identify strategies to enhance cell-based therapeutic efficacy. Here, we demonstrate the capacity for interferon (IFN)-γ licensing to enhance human MAPC efficacy and retention following early administration in a humanized mouse model of acute GvHD (aGvHD). Activation of the nuclear receptor peroxisome proliferator-activated receptor delta (PPARδ) negatively influenced the retention and efficacy of human MAPCs as well as IFN-γ-licensed MAPCs in the aGvHD model. PPARδ antagonism significantly enhanced the efficacy of human MAPCs when administered early in the humanized aGvHD model. COX-2 expression in human MAPC was significantly decreased in IFN-γ licensed MAPCs exposed to a PPARδ agonist. Importantly, MAPC exposure to the PPARδ antagonist in the presence of a COX-2 inhibitor indomethacin before administration significantly reduced the efficacy of PPARδ antagonized MAPCs in the aGvHD humanized mouse model. This is the first study to demonstrate the importance of PPARδ in human MAPC efficacy in vivo and highlights the importance of understanding the disease microenvironment in which cell-based therapies are to be administered. In particular, the presence of PPARδ ligands may negatively influence MAPC or MSC therapeutic efficacy.


Assuntos
Células-Tronco Adultas , Doença Enxerto-Hospedeiro , Células-Tronco Mesenquimais , PPAR delta , Animais , Doença Enxerto-Hospedeiro/terapia , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células-Tronco Multipotentes/metabolismo , PPAR delta/metabolismo
4.
Sci Rep ; 11(1): 13549, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34193955

RESUMO

Dysregulation of the immune system can initiate chronic inflammatory responses that exacerbate disease pathology. Multipotent adult progenitor cells (MAPC cells), an adult adherent bone-marrow derived stromal cell, have been observed to promote the resolution of uncontrolled inflammatory responses in a variety of clinical conditions including acute ischemic stroke, acute myocardial infarction (AMI), graft vs host disease (GvHD), and acute respiratory distress syndrome (ARDS). One of the proposed mechanisms by which MAPC cells modulate immune responses is via the induction of regulatory T cells (Tregs), however, the mechanism(s) involved remains to be fully elucidated. Herein, we demonstrate that, in an in vitro setting, MAPC cells increase Treg frequencies by promoting Treg proliferation and CD4+ T cell differentiation into Tregs. Moreover, MAPC cell-induced Tregs (miTregs) have a more suppressive phenotype characterized by increased expression of CTLA-4, HLA-DR, and PD-L1 and T cell suppression capacity. MAPC cells also promoted Treg activation by inducing CD45RA+ CD45RO+ transitional Tregs. Additionally, we identify transforming growth factor beta (TGFß) as an essential factor for Treg induction secreted by MAPC cells. Furthermore, inhibition of indoleamine 2, 3-dioxygenase (IDO) resulted in decreased Treg induction by MAPC cells demonstrating IDO involvement. Our studies also show that CD14+ monocytes play a critical role in Treg induction by MAPC cells. Our study describes MAPC cell dependent Treg phenotypic changes and provides evidence of potential mechanisms by which MAPC cells promote Treg differentiation.


Assuntos
Células-Tronco Adultas/imunologia , Tolerância Imunológica , Monócitos/imunologia , Células-Tronco Multipotentes/imunologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/imunologia , Humanos
5.
Wound Repair Regen ; 29(3): 380-392, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33655577

RESUMO

Stem cells have been shown to have potential as a new therapy for burns and promote wound healing through decreasing inflammation and increasing angiogenesis. Multipotent adult progenitor cells (MAPC® cells) are a subpopulation of bone marrow-derived stem cells with outstanding self-renewal and differentiation capacity. MAPC cells also secrete a wide range of cytokines which can affect cellular activities. This article aimed to examine the effects of MAPC cells treatment on burn injury repair using a mouse model of partial thickness burn injury. The immunomodulatory effect of MAPC cells was investigated in vitro using a simultaneous T-cell proliferation assay. Partial thickness burns were created on the dorsal surface of mice and MAPC cells were administered via intradermal injection to the wound margins 24 h post-burn injury. The burn tissues were analysed macroscopically to determine wound area and histologically assessed to determine wound width and rate of re-epithelialisation. Immunohistochemistry and ELISA were employed to assess cell proliferation, inflammation and angiogenesis and collagen deposition in the burn area. MAPC cells inhibit the proliferation of stimulated T cells in culture. Burns intradermally injected with MAPC cells showed a significant reduction in the macroscopic wound area, histologic wound width and had an increased rate of re-epithelialisation. Immunohistochemistry and ELISA analysis of burn tissues showed dampened inflammation evidenced by a reduction in neutrophilic infiltration and modulation of inflammatory cytokines. Angiogenesis within the burn area was also improved in MAPC cell treated mice. However, no significant effect of MAPC cell treatment was observed on extracellular matrix production. Treatment of burns with MAPC cells improved burn injury repair with reduced time to healing, decreased inflammation and increased angiogenesis. These findings demonstrate the promising effects of MAPC cells on burn injury repair and suggest MAPC cells as a candidate source for clinical cell therapies.


Assuntos
Queimaduras , Cicatrização , Animais , Queimaduras/terapia , Inflamação , Camundongos , Reepitelização , Células-Tronco
6.
Am J Transplant ; 21(4): 1402-1414, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32506663

RESUMO

Ex vivo normothermic machine perfusion (NMP) of donor kidneys prior to transplantation provides a platform for direct delivery of cellular therapeutics to optimize organ quality prior to transplantation. Multipotent Adult Progenitor Cells (MAPC® ) possess potent immunomodulatory properties that could minimize ischemia reperfusion injury. We investigated the potential capability of MAPC cells in kidney NMP. Pairs (5) of human kidneys, from the same donor, were simultaneously perfused for 7 hours. Kidneys were randomly allocated to receive MAPC treatment or control. Serial samples of perfusate, urine, and tissue biopsies were taken for comparison. MAPC-treated kidneys demonstrated improved urine output (P = .009), decreased expression of injury biomarker NGAL (P = .012), improved microvascular perfusion on contrast-enhanced ultrasound (cortex P = .019, medulla P = .001), downregulation of interleukin (IL)-1ß (P = .050), and upregulation of IL-10 (P < .047) and Indolamine-2, 3-dioxygenase (P = .050). A chemotaxis model demonstrated decreased neutrophil recruitment when stimulated with perfusate from MAPC-treated kidneys (P < .001). Immunofluorescence revealed prelabeled MAPC cells in the perivascular space of kidneys during NMP. We report the first successful delivery of cellular therapy to a human kidney during NMP. Kidneys treated with MAPC cells demonstrate improvement in clinically relevant parameters and injury biomarkers. This novel method of cell therapy delivery provides an exciting opportunity to recondition organs prior to transplantation.


Assuntos
Transplante de Rim , Traumatismo por Reperfusão , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Rim , Transplante de Rim/efeitos adversos , Preservação de Órgãos , Perfusão , Traumatismo por Reperfusão/prevenção & controle
7.
Biointerphases ; 15(6): 061015, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33356337

RESUMO

Cryopreservation is an essential part of tissue banking and effective cryopreservation methods are critical for the development of cost-effective cell therapy products. Cell sheets are an attractive subset of cell therapy types, and cryopreservation has the potential to further drive down costs of allogeneic cell sheet therapy. This is currently a challenge as adhered cell monolayers are more susceptible to membrane damage during the freezing process. In this article, we investigate the performance of a surface-modified dressing for the cryopreservation of cells and strategies to improve cell recovery. Cryopreservation of multipotent adult progenitor cells (MAPC®) was performed on cells following their attachment to a surface for different periods of time. MAPC cells, given just 1 h to attach, washed off and were not recovered on the surface following thawing. Cells attached for longer periods, elongated further, and were more susceptible to damage from cryopreservation. A temporal window was identified that could allow cryopreservation on adherent surfaces where cells had attached to a surface without full elongation. By functionalizing the surface with coupled hyaluronic acid, cell spreading was initially retarded, thereby widening this temporal window. This approach demonstrates a novel method for enhancing the recovery of cryopreserved cell sheets on surfaces.


Assuntos
Criopreservação/métodos , Ácido Hialurônico/química , Células-Tronco/citologia , Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Ácido Hialurônico/farmacologia , Polímeros/química , Células-Tronco/metabolismo , Propriedades de Superfície
8.
Stem Cell Res Ther ; 11(1): 299, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32680566

RESUMO

BACKGROUND: Stem cell therapies have been widely investigated for their healing effects. However, the translation of these therapies has been hampered by the requirement to deliver live allogeneic or autologous cells directly to the wound in a clinical setting. Multipotent adult progenitor cells (MAPC® cells) are a subpopulation of bone marrow-derived adherent stem cells that secrete a wide range of factors known to accelerate the wound healing process. The aim of this study was to determine the impact of MAPC cells secretome on healing outcomes without the presence of MAPC cells. METHODS: The effect of MAPC-conditioned medium (MAPC-CM) on the capacity of keratinocytes, fibroblasts and endothelial cells to migrate and proliferate was determined in vitro using scratch wound closure and WST1 assay, respectively. The effect of MAPC-CM on collagen deposition and angiogenesis was also assessed using in vitro methods. Additionally, two excisional wounds were created on the dorsal surface of mice (n = 8/group) and 100 µL of 20× MAPC-CM were intradermally injected to the wound margins. Wound tissues were collected at 3, 7 and 14 days post-wounding and stained with H&E for microscopic analysis. Immunohistochemistry was performed to investigate inflammation, angiogenesis and collagen deposition in the wounds. RESULTS: Skin fibroblasts, keratinocytes and endothelial cells treated with MAPC-CM all showed improved rates of scratch closure and increased cellular proliferation. Moreover, fibroblasts treated with MAPC-CM deposited more collagens I and III and endothelial cells treated with MAPC-CM showed increased capillary tube formation. Murine excisional wounds intradermally injected with MAPC-CM showed a significant reduction in the wound area and an increase in the rate of reepithelialisation. The results also showed that inflammatory cell infiltration was decreased while an increase in angiogenesis, as well as collagens I and III expressions, was observed. CONCLUSION: These findings suggest that factors produced by MAPC cells can have an important effect on cutaneous wound healing by affecting skin cell proliferation and migration, balancing inflammation and improving the formation of extracellular matrix and angiogenesis. Development of stem cell-free therapy for the treatment of wounds may be a more clinically translatable approach for improving healing outcomes.


Assuntos
Células Endoteliais , Cicatrização , Adulto , Animais , Meios de Cultivo Condicionados/farmacologia , Humanos , Inflamação , Camundongos , Células-Tronco Multipotentes , Pele
9.
Front Immunol ; 11: 1226, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32714318

RESUMO

Background: Pre-clinical research with multi-potent adult progenitor cells (MAPC® cells, Multistem, Athersys Inc., Cleveland, Ohio) suggests their potential as an anti-inflammatory and immunomodulatory therapy in organ transplantation. Normothermic machine perfusion of the liver (NMP-L) has been proposed as a way of introducing therapeutic agents into the donor organ. Delivery of cellular therapy to human donor livers using this technique has not yet been described in the literature. The primary objectives of this study were to develop a technique for delivering cellular therapy to human donor livers using NMP-L and demonstrate engraftment. Methods: Six discarded human livers were perfused for 6 h at 37°C using the Liver Assist (Organ Assist, Groningen). 50 × 106 CMPTX-labeled MAPC cells were infused directly into the right lobe via the hepatic artery (HA, n = 3) or portal vein (PV, n = 3) over 20 min at different time points during the perfusion. Perfusion parameters were recorded and central and peripheral biopsies were taken at multiple time-points from both lobes and subjected to standard histological stains and confocal microscopy. Perfusate was analyzed using a 35-plex multiplex assay and proteomic analysis. Results: There was no detrimental effect on perfusion flow parameters on infusion of MAPC cells by either route. Three out of six livers met established criteria for organ viability. Confocal microscopy demonstrated engraftment of MAPC cells across vascular endothelium when perfused via the artery. 35-plex multiplex analysis of perfusate yielded 13 positive targets, 9 of which appeared to be related to the infusion of MAPC cells (including Interleukin's 1b, 4, 5, 6, 8, 10, MCP-1, GM-CSF, SDF-1a). Proteomic analysis revealed 295 unique proteins in the perfusate from time-points following the infusion of cellular therapy, many of which have strong links to MAPC cells and mesenchymal stem cells in the literature. Functional enrichment analysis demonstrated their immunomodulatory potential. Conclusion: We have demonstrated that cells can be delivered directly to the target organ, prior to host immune cell population exposure and without compromising the perfusion. Transendothelial migration occurs following arterial infusion. MAPC cells appear to secrete a host of soluble factors that would have anti-inflammatory and immunomodulatory benefits in a human model of liver transplantation.


Assuntos
Células-Tronco Adultas , Transplante de Fígado , Doadores Vivos , Transplante de Células-Tronco , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Biomarcadores , Terapia Baseada em Transplante de Células e Tecidos , Quimiocinas/metabolismo , Terapia Combinada , Citocinas/metabolismo , Imunofluorescência , Humanos , Imuno-Histoquímica , Imunofenotipagem , Imunoterapia , Transplante de Fígado/métodos , Preservação de Órgãos/métodos , Perfusão , Proteoma , Transplante de Células-Tronco/métodos
10.
Front Immunol ; 9: 645, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29740426

RESUMO

Lymphodepletion strategies are used in the setting of transplantation (including bone marrow, hematopoietic cell, and solid organ) to create space or to prevent allograft rejection and graft versus host disease. Following lymphodepletion, there is an excess of IL-7 available, and T cells that escape depletion respond to this cytokine undergoing accelerated proliferation. Moreover, this environment promotes the skew of T cells to a Th1 pro-inflammatory phenotype. Existing immunosuppressive regimens fail to control this homeostatic proliferative (HP) response, and thus the development of strategies to successfully control HP while sparing T cell reconstitution (providing a functioning immune system) represents a significant unmet need in patients requiring lymphodepletion. Multipotent adult progenitor cells (MAPC®) have the capacity to control T cell proliferation and Th1 cytokine production. Herein, this study shows that MAPC cells suppressed anti-thymocyte globulin-induced cytokine production but spared T cell reconstitution in a pre-clinical model of lymphodepletion. Importantly, MAPC cells administered intraperitoneally were efficacious in suppressing interferon-γ production and in promoting the expansion of regulatory T cells in the lymph nodes. MAPC cells administered intraperitoneally accumulated in the omentum but were not present in the spleen suggesting a role for soluble factors. MAPC cells suppressed lymphopenia-induced cytokine production in a prostaglandin E2-dependent manner. This study suggests that MAPC cell therapy may be useful as a novel strategy to target lymphopenia-induced pathogenic T cell responses in lymphodepleted patients.


Assuntos
Células-Tronco Adultas/imunologia , Rejeição de Enxerto/prevenção & controle , Imunoterapia/métodos , Células-Tronco Pluripotentes/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Transplante , Células-Tronco Adultas/ultraestrutura , Animais , Proliferação de Células , Células Cultivadas , Dinoprostona/metabolismo , Modelos Animais de Doenças , Homeostase , Humanos , Ativação Linfocitária , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/transplante
11.
Stem Cells Transl Med ; 5(5): 628-31, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27025689

RESUMO

UNLABELLED: Cell-based therapy has potential therapeutic value in autoimmune diseases such as rheumatoid arthritis (RA). In RA, reduction of disease activity has been associated with improvement in the function of regulatory T cells (Treg) and attenuated responses of proinflammatory effector T cells (Teff). Mesenchymal stem cells (MSCs) and related multipotent adult progenitor cells (MAPC) have strong anti-inflammatory and immunomodulatory properties and may be able to "reset" the immune system to a pre-RA state. MAPC are MSC-like cells that are slightly earlier in lineage, have greater expansion capacity, and can be used as "off-the-shelf" therapy. Assessment of cell-based therapy to treat arthritis and related diseases is limited by the lack of available biological correlates that can be measured early on and indicate treatment response. We set out to develop a functional measure that could be used ex vivo as a biomarker of response. We were able to demonstrate that MAPC products could inhibit Teff responses from patients with active RA and that Treg from RA patients suppressed Teff. This assay used ex vivo can be used with MAPC or Treg alone or in combination and reflects the overall level of Teff suppression. Use of a novel functional biomarker as an exploratory endpoint in trials of cell-based therapy should be of value to detect biological outcomes at a point prior to the time that clinical response might be observed. SIGNIFICANCE: Therapy with mesenchymal stem cells and related multipotent adult progenitor cells is immune modifying in a variety of diseases. There is interest in using cell-based therapy in rheumatoid arthritis (RA) to induce tolerance and "reset" the immune system to its pre-RA state. In a clinical trial, it should be known as soon as possible if there is a chance of response. A biomarker has been developed that permits measurement of the effects of cell-based therapy on effector T cell function.


Assuntos
Células-Tronco Adultas/metabolismo , Artrite Reumatoide/terapia , Bioensaio/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Multipotentes/metabolismo , Linfócitos T Reguladores/metabolismo , Células-Tronco Adultas/imunologia , Artrite Reumatoide/sangue , Artrite Reumatoide/diagnóstico , Artrite Reumatoide/imunologia , Biomarcadores/sangue , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Humanos , Ativação Linfocitária , Células-Tronco Multipotentes/imunologia , Fenótipo , Valor Preditivo dos Testes , Linfócitos T Reguladores/imunologia , Resultado do Tratamento
12.
Mol Ther ; 23(11): 1783-1793, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26216515

RESUMO

T-cell depletion therapy is used to prevent acute allograft rejection, treat autoimmunity and create space for bone marrow or hematopoietic cell transplantation. The evolved response to T-cell loss is a transient increase in IL-7 that drives compensatory homeostatic proliferation (HP) of mature T cells. Paradoxically, the exaggerated form of this process that occurs following lymphodepletion expands effector T-cells, often causing loss of immunological tolerance that results in rapid graft rejection, autoimmunity, and exacerbated graft-versus-host disease (GVHD). While standard immune suppression is unable to treat these pathologies, growing evidence suggests that manipulating the incipient process of HP increases allograft survival, prevents autoimmunity, and markedly reduces GVHD. Multipotent adult progenitor cells (MAPC) are a clinical grade immunomodulatory cell therapy known to alter γ-chain cytokine responses in T-cells. Herein, we demonstrate that MAPC regulate HP of human T-cells, prevent the expansion of Th1, Th17, and Th22 effectors, and block the development of pathogenic allograft responses. This occurs via IL-1ß-primed secretion of PGE2 and activates T-cell intrinsic regulatory mechanisms (SOCS2, GADD45A). These data provide proof-of-principle that HP of human T-cells can be targeted by cellular and molecular therapies and lays a basis for the development of novel strategies to prevent immunopathology in lymphodepleted patients.


Assuntos
Células-Tronco Adultas/fisiologia , Dinoprostona/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Interleucina-7/imunologia , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Multipotentes/fisiologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Adulto , Células-Tronco Adultas/imunologia , Autoimunidade , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Rejeição de Enxerto , Humanos , Tolerância Imunológica , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Interleucina-7/metabolismo , Depleção Linfocítica/efeitos adversos , Masculino , Células-Tronco Mesenquimais/imunologia , Células-Tronco Multipotentes/imunologia , Proteínas Nucleares/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Transplante Homólogo/métodos , Adulto Jovem
13.
Transplant Res ; 3(1): 19, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25671090

RESUMO

BACKGROUND: Primary graft dysfunction (PGD) is a significant cause of early morbidity and mortality following lung transplantation. Improved organ preservation techniques will decrease ischemia-reperfusion injury (IRI) contributing to PGD. Adult bone marrow-derived adherent stem cells, including mesenchymal stromal (stem) cells (MSCs) and multipotent adult progenitor cells (MAPCs), have potent anti-inflammatory actions, and we thus postulated that intratracheal MAPC administration during donor lung processing would decrease IRI. The goal of the study was therefore to determine if intratracheal MAPC instillation would decrease lung injury and inflammation in an ex vivo human lung explant model of prolonged cold storage and subsequent reperfusion. METHODS: Four donor lungs not utilized for transplant underwent 8 h of cold storage (4°C). Following rewarming for approximately 30 min, non-HLA-matched allogeneic MAPCs (1 × 10(7) MAPCs/lung) were bronchoscopically instilled into the left lower lobe (LLL) and vehicle comparably instilled into the right lower lobe (RLL). The lungs were then perfused and mechanically ventilated for 4 h and subsequently assessed for histologic injury and for inflammatory markers in bronchoalveolar lavage fluid (BALF) and lung tissue. RESULTS: All LLLs consistently demonstrated a significant decrease in histologic and BALF inflammation compared to vehicle-treated RLLs. CONCLUSIONS: These initial pilot studies suggest that use of non-HLA-matched allogeneic MAPCs during donor lung processing can decrease markers of cold ischemia-induced lung injury.

14.
J Immunol ; 190(9): 4542-52, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23547116

RESUMO

A major goal of immunotherapy remains the control of pathogenic T cell responses that drive autoimmunity and allograft rejection. Adherent progenitor cells, including mesenchymal stromal cells (MSCs) and multipotent adult progenitor cells (MAPCs), represent attractive immunomodulatory cell therapy candidates currently active in clinical trials. MAPCs can be distinguished from MSCs on the basis of cellular phenotype, size, transcriptional profile, and expansion capacity. However, despite their ongoing evaluation in autoimmune and allogeneic solid organ transplantation settings, data supporting the immune regulatory potential of clinical-grade MAPCs are limited. In this study, we used allogeneic islet transplantation as a model indication to assess the ability of clinical-grade MAPCs to control T cell responses that drive immunopathology in human autoimmune disease and allograft rejection. MAPCs suppressed T cell proliferation and Th1 and Th17 cytokine production while increasing secretion of IL-10 and were able to suppress effector functions of bona fide autoreactive T cells from individuals with type 1 diabetes mellitus, including killing of human islets. Furthermore, MAPCs favored the proliferation of regulatory T cells during homeostatic expansion driven by γ-chain cytokines and exerted a durable, yet reversible, control of T cell function. MAPC suppression required licensing and proceeded via IDO-mediated tryptophan catabolism. Therefore, the common immune modulatory characteristics of clinical-grade MAPCs shown in this study suggest that they can be regarded as an alternative source of adult progenitor cells with similar clinical usefulness to MSCs. Taken collectively, these findings may guide the successful deployment of both MSCs and MAPCs for the amelioration of human autoimmunity and allograft rejection.


Assuntos
Autoimunidade/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Ativação Linfocitária/imunologia , Células-Tronco/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Adulto , Células-Tronco Adultas/imunologia , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Tipo 1/imunologia , Rejeição de Enxerto/imunologia , Humanos , Imunomodulação/imunologia , Interleucina-10/imunologia , Masculino , Triptofano/imunologia , Adulto Jovem
15.
Curr Opin Organ Transplant ; 17(6): 675-80, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23111647

RESUMO

PURPOSE OF REVIEW: Stem cell therapy for the treatment of cardiovascular disease is rapidly moving from bench to bedside. In the settings of acute and chronic myocardial injury, approaches to treatment have explored various cell populations, delivery methods, and times of administration. RECENT FINDINGS: Although initial studies in patients were performed with unfractionated bone marrow cells, further investigations in animal models of myocardial disease have elucidated mechanisms of benefit and opened doors to treatment strategies with stem cells of varied derivation. SUMMARY: Allogeneic stem cell populations have demonstrated therapeutic promise in ischemic heart disease, without a requirement for immunosuppressive drugs, and offer the potential to create large-scale, cryopreserved banks of cells for 'off the shelf' utility.


Assuntos
Transplante de Medula Óssea , Transplante de Células-Tronco Mesenquimais , Isquemia Miocárdica/cirurgia , Animais , Células da Medula Óssea , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos , Isquemia Miocárdica/fisiopatologia , Transplante de Células-Tronco/métodos , Transplante Autólogo , Transplante Homólogo , Resultado do Tratamento
16.
Cytotherapy ; 14(8): 994-1004, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22687190

RESUMO

BACKGROUND AIMS: Clinical results from acute myocardial infarction (AMI) patients treated with MultiStem®, a large-scale expanded adherent multipotent progenitor cell population (MAPC), have demonstrated a strong safety and benefit profile for these cells. The mechanism of benefit with MAPC treatment is a result, in part, of its ability to induce neovascularization through trophic support. Production of clinical-grade stem cell products requires the development of lot-release criteria based on potency assays that directly reflect the fundamental mechanistic pathway underlying the therapeutic response to verify manufacturing process consistency and product potency. METHODS AND RESULTS: Using an in vitro endothelial tube formation assay, a potency assay has been developed that reflects MAPC pro-angiogenic activity. Serum-free conditioned media collected from MAPC culture induced endothelial tube formation. A proteomic survey of angiogenic factors produced by the cells in vitro revealed candidate factors linked to angiogenic potency. Three cytokines, chemokine (C-X-C motif) ligand 5 (CXCL5), interleukin 8 (IL-8) and vascular endothelial growth factor (VEGF), were required for this angiogenic activity. Depletion of any of these factors from the media prevented tube formation, while adding back increasing amounts of these cytokines into the depleted serum-free conditioned media established the lower limits of each of the cytokines required to induce angiogenesis. CONCLUSIONS: A necessary threshold of angiogenic factor expression was established using an in vitro angiogenesis assay. By correlating the levels of the cytokines required to induce tube formation in vitro with levels of the factors found in the spent media from manufacturing production runs, detection of these factors was identified as a surrogate potency assay with defined pass/fail criteria.


Assuntos
Técnicas de Cultura de Células , Terapia Baseada em Transplante de Células e Tecidos , Células Endoteliais/citologia , Células-Tronco Multipotentes/citologia , Neovascularização Fisiológica , Células da Medula Óssea/citologia , Diferenciação Celular , Meios de Cultivo Condicionados , Meios de Cultura Livres de Soro , Citocinas/metabolismo , Expressão Gênica , Humanos , Interleucina-8/metabolismo , Células-Tronco Multipotentes/transplante , Infarto do Miocárdio/terapia , Receptores Acoplados a Proteínas G/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Cell Transplant ; 21(6): 1109-20, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22004910

RESUMO

Acute myocardial infarction (AMI) results in ischemic damage and death of cardiomyocytes and loss of vasculature. Stem cell therapy has emerged as a potentially promising strategy for maximizing cardiac function following ischemic injury. Issues of cell source, delivery, and quantification of response have challenged development of clinically viable strategies. In this study we investigate the effects of a well-defined bone marrow-derived allogeneic cell product delivered by catheter directly to the myocardium via the infarct-related vessel on global and regional measures of left ventricular (LV) function in a porcine model of anterior wall myocardial infarction. Multipotent adult progenitor cells (MAPCs) were derived and expanded from the bone marrow of a donor Yorkshire pig. Anterior wall myocardial infarction (AMI) was induced by 90 min of mid-LAD occlusion using a balloon catheter. Two days after AMI was induced, either vehicle (Plasma Lyte-A, n = 7), low-dose (20 million, n = 6), or high-dose (200 million, n = 6) MAPCs were delivered directly to the myocardium via the infarct-related vessel using a transarterial microsyringe catheter-based delivery system. Echocardiography was used to measure LV function as a function of time after AMI. Animals that received low-dose cell treatment showed significant improvement in regional and global LV function and remodeling compared to the high-dose or control animals. Direct myocardial delivery of allogeneic MAPCs 2 days following AMI through the vessel wall of the infarct-related vessel is safe and results in delivery of cells throughout the infarct zone and improved cardiac function despite lack of long-term cell survival. These data further support the hypothesis of cell-based myocardial tissue repair by a paracrine mechanism and suggest a clinically translatable strategy for delivering cells at any time after AMI to modulate cardiac remodeling and function.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Multipotentes/citologia , Infarto do Miocárdio/terapia , Função Ventricular Esquerda/fisiologia , Doença Aguda , Animais , Cateterismo , Células Cultivadas , Modelos Animais de Doenças , Ecocardiografia , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/transplante , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Suínos , Transplante Homólogo , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Circ Res ; 110(2): 304-11, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22052917

RESUMO

RATIONALE: MultiStem is an allogeneic bone marrow-derived adherent adult stem cell product that has shown efficacy in preclinical models of acute myocardial infarction (AMI). In this phase I clinical trial in patients with first ST-elevation-myocardial infarction (STEMI), we combine first-in-man delivery of MultiStem with a first-in-coronary adventitial delivery system to determine the effects of this system on left ventricular function at 4 months after AMI. OBJECTIVE: Test the effects of adventitial delivery of Multistem in the peri-infarct period in patients with first STEMI. METHODS AND RESULTS: This study was a phase I, open-label, dose-escalating registry control group study. Nineteen patients received MultiStem (20 million, n=6; 50 million, n=7; or 100 million, n=6) and 6 subjects were assigned to the registry control group. Two to 5 days after AMI, we delivered MultiStem to the adventitia of the infarct-related vessel in patients with first-time STEMI. All patients underwent primary percutaneous coronary intervention with resulting Thrombolysis In Myocardial Infarction grade 3 flow and with ejection fraction (EF) ≤45% as determined by echocardiogram or left ventriculogram within 12 hours of primary percutaneous coronary intervention. The cell product (20 million, 50 million, or 100 million) was well tolerated, and no serious adverse events were deemed related to MultiStem. There was no increase in creatine kinase-MB or troponin associated with the adventitial delivery of MultiStem. In patients with EF determined to be ≤45% by a core laboratory within 24 hours before the MultiStem injection, we observed a 0.9 (n=4), 3.9 (n=4), 13.5 (n=5), and 10.9 (n=2) percent absolute increases in EF in the registry, 20 million, 50 million, and 100 million dose groups, respectively. The increases in EF in the 50 million and 100 million groups were accompanied by 25.4 and 8.4 mL increases in left ventricular stroke volume. CONCLUSIONS: In this study, the delivery of MultiStem to the myocardium in patients with recent STEMI was well tolerated and safe. In patients who exhibited significant myocardial damage, the delivery of ≥50 million MultiStem resulted in improved EF and stroke volume 4 months later. These findings support further development of MultiStem in patients with AMI and they validate the potential of a system for delivery of adult stem cells at any time after primary percutaneous coronary intervention.


Assuntos
Células-Tronco Adultas/transplante , Angioplastia Coronária com Balão , Transplante de Medula Óssea , Infarto do Miocárdio/terapia , Células-Tronco Adultas/imunologia , Idoso , Transplante de Medula Óssea/efeitos adversos , Terapia Combinada , Tecido Conjuntivo , Feminino , Humanos , Masculino , Microinjeções , Pessoa de Meia-Idade , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Miocárdio/patologia , Recuperação de Função Fisiológica , Sistema de Registros , Volume Sistólico , Fatores de Tempo , Transplante Homólogo , Resultado do Tratamento , Estados Unidos , Função Ventricular Esquerda
19.
J Neurosci ; 31(3): 944-53, 2011 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-21248119

RESUMO

Macrophage-mediated axonal dieback presents an additional challenge to regenerating axons after spinal cord injury. Adult adherent stem cells are known to have immunomodulatory capabilities, but their potential to ameliorate this detrimental inflammation-related process has not been investigated. Using an in vitro model of axonal dieback as well as an adult rat dorsal column crush model of spinal cord injury, we found that multipotent adult progenitor cells (MAPCs) can affect both macrophages and dystrophic neurons simultaneously. MAPCs significantly decrease MMP-9 (matrix metalloproteinase-9) release from macrophages, effectively preventing induction of axonal dieback. MAPCs also induce a shift in macrophages from an M1, or "classically activated" proinflammatory state, to an M2, or "alternatively activated" antiinflammatory state. In addition to these effects on macrophages, MAPCs promote sensory neurite outgrowth, induce sprouting, and further enable axons to overcome the negative effects of macrophages as well as inhibitory proteoglycans in their environment by increasing their intrinsic growth capacity. Our results demonstrate that MAPCs have therapeutic benefits after spinal cord injury and provide specific evidence that adult stem cells exert positive immunomodulatory and neurotrophic influences.


Assuntos
Axônios/fisiologia , Macrófagos/fisiologia , Células-Tronco Multipotentes/fisiologia , Regeneração Nervosa/fisiologia , Células do Corno Posterior/fisiologia , Traumatismos da Medula Espinal/metabolismo , Animais , Western Blotting , Células Cultivadas , Imuno-Histoquímica , Macrófagos/citologia , Metaloproteinase 9 da Matriz/metabolismo , Compressão Nervosa , Células do Corno Posterior/citologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Traumatismos da Medula Espinal/fisiopatologia
20.
Cell Med ; 2(2): 43-53, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-26998401

RESUMO

Type 1 diabetes is an autoimmune disorder that leads to destruction of pancreatic ß islet cells and is a growing global health issue. While insulin replacement remains the standard therapy for type 1 diabetes, exogenous insulin does not mimic the physiology of insulin secretion. Transplantation of pancreatic islets has the potential to cure this disease; however, there are several major limitations to widespread implementation of islet transplants. The use of mesenchymal stromal cells (MSCs) in the treatment of type 1 diabetes has been investigated as an adjunct therapy during islet graft administration to prevent initial islet loss and promote engraftment and revascularization of islets. In this review we will discuss the results of recent MSC studies in animal models of diabetes with a focus on islet transplantation and explore the potential for these findings to be extended to clinical use for the treatment of type 1 diabetes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA