Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Rev ; 74(3): 462-505, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35710133

RESUMEN

The concept of local formation of angiotensin II in the kidney has changed over the last 10-15 years. Local synthesis of angiotensinogen in the proximal tubule has been proposed, combined with prorenin synthesis in the collecting duct. Binding of prorenin via the so-called (pro)renin receptor has been introduced, as well as megalin-mediated uptake of filtered plasma-derived renin-angiotensin system (RAS) components. Moreover, angiotensin metabolites other than angiotensin II [notably angiotensin-(1-7)] exist, and angiotensins exert their effects via three different receptors, of which angiotensin II type 2 and Mas receptors are considered renoprotective, possibly in a sex-specific manner, whereas angiotensin II type 1 (AT1) receptors are believed to be deleterious. Additionally, internalized angiotensin II may stimulate intracellular receptors. Angiotensin-converting enzyme 2 (ACE2) not only generates angiotensin-(1-7) but also acts as coronavirus receptor. Multiple, if not all, cardiovascular diseases involve the kidney RAS, with renal AT1 receptors often being claimed to exert a crucial role. Urinary RAS component levels, depending on filtration, reabsorption, and local release, are believed to reflect renal RAS activity. Finally, both existing drugs (RAS inhibitors, cyclooxygenase inhibitors) and novel drugs (angiotensin receptor/neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors, soluble ACE2) affect renal angiotensin formation, thereby displaying cardiovascular efficacy. Particular in the case of the latter three, an important question is to what degree they induce renoprotection (e.g., in a renal RAS-dependent manner). This review provides a unifying view, explaining not only how kidney angiotensin formation occurs and how it is affected by drugs but also why drugs are renoprotective when altering the renal RAS. SIGNIFICANCE STATEMENT: Angiotensin formation in the kidney is widely accepted but little understood, and multiple, often contrasting concepts have been put forward over the last two decades. This paper offers a unifying view, simultaneously explaining how existing and novel drugs exert renoprotection by interfering with kidney angiotensin formation.


Asunto(s)
Angiotensinógeno , Enfermedades Cardiovasculares , Femenino , Humanos , Masculino , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Angiotensinógeno/metabolismo , Enfermedades Cardiovasculares/metabolismo , Sistemas de Liberación de Medicamentos , Riñón/irrigación sanguínea , Riñón/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina , Inhibidores del Cotransportador de Sodio-Glucosa 2/metabolismo
2.
Am J Transplant ; 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38447886

RESUMEN

The extent to which tissue-resident memory T (TRM) cells in transplanted organs possess alloreactivity is uncertain. This study investigates the alloreactive potential of TRM cells in kidney explants from 4 patients who experienced severe acute rejection leading to graft loss. Alloreactive T cell receptor (TCR) clones were identified in pretransplant blood samples through mixed lymphocyte reactions, followed by single-cell RNA and TCR sequencing of the proliferated recipient T cells. Subsequently, these TCR clones were traced in the TRM cells of kidney explants, which were also subjected to single-cell RNA and TCR sequencing. The proportion of recipient-derived TRM cells expressing an alloreactive TCR in the 4 kidney explants varied from 0% to 9%. Notably, these alloreactive TCRs were predominantly found among CD4+ and CD8+ TRM cells with an effector phenotype. Intriguingly, these clones were present not only in recipient-derived TRM cells but also in donor-derived TRM cells, constituting up to 4% of the donor population, suggesting the presence of self-reactive TRM cells. Overall, our study demonstrates that T cells with alloreactive potential present in the peripheral blood prior to transplantation can infiltrate the kidney transplant and adopt a TRM phenotype.

3.
Transpl Int ; 37: 12468, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38699175

RESUMEN

Kidney organoids are an innovative tool in transplantation research. The aim of the present study was to investigate whether kidney organoids are susceptible for allo-immune attack and whether they can be used as a model to study allo-immunity in kidney transplantation. Human induced pluripotent stem cell-derived kidney organoids were co-cultured with human peripheral blood mononuclear cells (PBMC), which resulted in invasion of allogeneic T-cells around nephron structures and macrophages in the stromal cell compartment of the organoids. This process was associated with the induction of fibrosis. Subcutaneous implantation of kidney organoids in immune-deficient mice followed by adoptive transfer of human PBMC led to the invasion of diverse T-cell subsets. Single cell transcriptomic analysis revealed that stromal cells in the organoids upregulated expression of immune response genes upon immune cell invasion. Moreover, immune regulatory PD-L1 protein was elevated in epithelial cells while genes related to nephron differentiation and function were downregulated. This study characterized the interaction between immune cells and kidney organoids, which will advance the use of kidney organoids for transplantation research.


Asunto(s)
Trasplante de Riñón , Riñón , Organoides , Humanos , Organoides/inmunología , Animales , Riñón/inmunología , Ratones , Técnicas de Cocultivo , Leucocitos Mononucleares/inmunología , Células Madre Pluripotentes Inducidas/citología , Linfocitos T/inmunología , Sistema Inmunológico , Antígeno B7-H1/metabolismo , Macrófagos/inmunología
4.
Stem Cells ; 40(6): 577-591, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35524742

RESUMEN

Induced pluripotent stem cell (iPSC)-derived kidney organoids are a potential tool for the regeneration of kidney tissue. They represent an early stage of nephrogenesis and have been shown to successfsully vascularize and mature further in vivo. However, there are concerns regarding the long-term safety and stability of iPSC derivatives. Specifically, the potential for tumorigenesis may impede the road to clinical application. To study safety and stability of kidney organoids, we analyzed their potential for malignant transformation in a teratoma assay and following long-term subcutaneous implantation in an immune-deficient mouse model. We did not detect fully functional residual iPSCs in the kidney organoids as analyzed by gene expression analysis, single-cell sequencing and immunohistochemistry. Accordingly, kidney organoids failed to form teratoma. Upon long-term subcutaneous implantation of whole organoids in immunodeficient IL2Ry-/-RAG2-/- mice, we observed tumor formation in 5 out of 103 implanted kidney organoids. These tumors were composed of WT1+CD56+ immature blastemal cells and showed histological resemblance with Wilms tumor. No genetic changes were identified that contributed to the occurrence of tumorigenic cells within the kidney organoids. However, assessment of epigenetic changes revealed a unique cluster of differentially methylated genes that were also present in undifferentiated iPSCs. We discovered that kidney organoids have the capacity to form tumors upon long-term implantation. The presence of epigenetic modifications combined with the lack of environmental cues may have caused an arrest in terminal differentiation. Our results indicate that the safe implementation of kidney organoids should exclude the presence of pro-tumorigenic methylation in kidney organoids.


Asunto(s)
Células Madre Pluripotentes Inducidas , Teratoma , Animales , Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/patología , Ratones , Organogénesis , Organoides/metabolismo , Teratoma/patología
5.
Am J Transplant ; 22(12): 2723-2739, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35896477

RESUMEN

The use of extended criteria donor grafts is a promising strategy to increase the number of organ transplantations and reduce waitlist mortality. However, these organs are often compromised and/or damaged, are more susceptible to preservation injury, and are at risk for developing post-transplant complications. Ex vivo organ perfusion is a novel technology to preserve donor organs while providing oxygen and nutrients at distinct perfusion temperatures. This preservation method allows to resuscitate grafts and optimize function with therapeutic interventions prior to solid organ transplantation. Stem cell-based therapies are increasingly explored for their ability to promote regeneration and reduce the inflammatory response associated with in vivo reperfusion. The aim of this review is to describe the current state of stem cell-based therapies during ex vivo organ perfusion for the kidney, liver, lung, and heart. We discuss different strategies, including type of cells, route of administration, mechanisms of action, efficacy, and safety. The progress made within lung transplantation justifies the initiation of clinical trials, whereas more research is likely required for the kidney, liver, and heart to progress into clinical application. We emphasize the need for standardization of methodology to increase comparability between future (clinical) studies.


Asunto(s)
Trasplante de Órganos , Daño por Reperfusión , Humanos , Preservación de Órganos/métodos , Perfusión/métodos , Circulación Extracorporea , Células Madre
6.
Kidney Int ; 99(1): 134-147, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32918942

RESUMEN

Renin production by the kidney is of vital importance for salt, volume, and blood pressure homeostasis. The lack of human models hampers investigation into the regulation of renin and its relevance for kidney physiology. To develop such a model, we used human induced pluripotent stem cell-derived kidney organoids to study the role of renin and the renin-angiotensin system in the kidney. Extensive characterization of the kidney organoids revealed kidney-specific cell populations consisting of podocytes, proximal and distal tubular cells, stromal cells and endothelial cells. We examined the presence of various components of the renin-angiotensin system such as angiotensin II receptors, angiotensinogen, and angiotensin-converting enzymes 1 and 2. We identified by single-cell sequencing, immunohistochemistry, and functional assays that cyclic AMP stimulation induces a subset of pericytes to increase the synthesis and secretion of enzymatically active renin. Renin production by the organoids was responsive to regulation by parathyroid hormone. Subcutaneously implanted kidney organoids in immunodeficient IL2Ry-/-Rag2-/- mice were successfully vascularized, maintained tubular and glomerular structures, and retained capacity to produce renin two months after implantation. Thus, our results demonstrate that kidney organoids express renin and provide insights into the endocrine potential of human kidney organoids, which is important for regenerative medicine in the context of the endocrine system.


Asunto(s)
Células Madre Pluripotentes Inducidas , Renina , Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Animales , Células Endoteliales/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/metabolismo , Ratones , Organoides/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina
7.
Am J Transplant ; 21(7): 2348-2359, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33382194

RESUMEN

Normothermic machine perfusion (NMP) of injured kidneys offers the opportunity for interventions to metabolically active organs prior to transplantation. Mesenchymal stromal cells (MSCs) can exert regenerative and anti-inflammatory effects in ischemia-reperfusion injury. The aims of this study were to evaluate the safety and feasibility of MSC treatment of kidneys during NMP using a porcine autotransplantation model, and examine potential MSC treatment-associated kidney improvements up to 14 days posttransplant. After 75 min of kidney warm ischemia, four experimental groups of n = 7 underwent 14 h of oxygenated hypothermic machine perfusion. In three groups this was followed by 240 min of NMP with infusion of vehicle, 10 million porcine, or 10 million human adipose-derived MSCs. All kidneys were autotransplanted after contralateral nephrectomy. MSC treatment did not affect perfusion hemodynamics during NMP or cause adverse effects at reperfusion, with 100% animal survival. MSCs did not affect plasma creatinine, glomerular filtration rate, neutrophil gelatinase-associated lipocalin concentrations or kidney damage assessed by histology during the 14 days, and MSCs retention was demonstrated in renal cortex. Infusing MSCs during ex vivo NMP of porcine kidneys was safe and feasible. Within the short posttransplant follow-up period, no beneficial effects of ex vivo MSC therapy could be demonstrated.


Asunto(s)
Células Madre Mesenquimatosas , Preservación de Órganos , Animales , Humanos , Riñón , Perfusión , Porcinos , Trasplante Autólogo
8.
Stem Cells ; 38(6): 797-807, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32101344

RESUMEN

Mesenchymal stem cells (MSCs) are used in various clinical and preclinical models for immunomodulation. However, it remains unclear how the immunomodulatory effect of MSC is communicated. MSC-induced immunomodulation is known to be mediated through both MSC-secreted cytokines and direct cell-cell interactions. Recently, it has been demonstrated that metabolically inactive, heat-inactivated MSCs (HI-MSCs) have similar anti-inflammatory capacities in LPS-induced sepsis compared with viable MSC. To further investigate the immunomodulatory effects of MSC, we introduced MSC and HI-MSC in two animal models with different immunological causes. In the first model, allogeneic hearts were transplanted from C57BL/6 mice to BALB/c recipients. MSC in combination with mycophenolate mofetil (MMF) significantly improved graft survival compared with MMF alone, whereas the application of HI-MSC had no effect on graft survival. We revealed that control MSC dose-dependently inhibited CD3+ and CD8+ T-cell proliferation in vitro, whereas HI-MSC had no effect. In the second model, sepsis was induced in mice via cecal ligation and puncture. HI-MSC treatment significantly improved the overall survival, whereas control MSCs had no effect. in vitro studies demonstrated that HI-MSCs are more effectively phagocytosed by monocytes than control MSCs and induced cell death in particular of activated CD16+ monocytes, which may explain the immune protective effect of HI-MSC in the sepsis model. The results of our study demonstrate that MSC-mediated immunomodulation in sepsis is dependent on a passive recognition of MSC by monocytes, whereas fully functional MSCs are required for inhibition of T-cell-mediated allograft rejection.


Asunto(s)
Trasplante de Corazón/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Sepsis/etiología , Trasplante Homólogo/métodos , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Sepsis/patología
9.
Transpl Int ; 34(2): 233-244, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33207013

RESUMEN

Cellular therapy is a promising tool for improving the outcome of organ transplantation. Various cell types with different immunoregulatory and regenerative properties may find application for specific transplant rejection or injury-related indications. The current era is crucial for the development of cellular therapies. Preclinical models have demonstrated the feasibility of efficacious cell therapy in transplantation, early clinical trials have shown safety of several of these therapies, and the first steps towards efficacy studies in humans have been made. In this review, we address the current state of the art of cellular therapies in clinical transplantation and discuss monitoring tools and endpoints for these studies.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Trasplante de Órganos , Tratamiento Basado en Trasplante de Células y Tejidos , Rechazo de Injerto , Humanos
10.
Transpl Int ; 34(11): 2006-2018, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34459040

RESUMEN

The future clinical application of animal-to-human transplantation (xenotransplantation) is of importance to society as a whole. Favourable preclinical data relevant to cell, tissue and solid organ xenotransplants have been obtained from many animal models utilizing genetic engineering and protocols of pathogen-free husbandry. Findings have reached a tipping point, and xenotransplantation of solid organs is approaching clinical evaluation, the process of which now requires close deliberation. Such discussions include considering when there is sufficient evidence from preclinical animal studies to start first-in-human xenotransplantation trials. The present article is based on evidence and opinions formulated by members of the European Society for Organ Transplantation who are involved in the Transplantation Learning Journey project. The article includes a brief overview of preclinical concepts and biology of solid organ xenotransplantation, discusses the selection of candidates for first-in-human studies and considers requirements for study design and conduct. In addition, the paper emphasizes the need for a regulatory framework for xenotransplantation of solid organs and the essential requirement for input from public and patient stakeholders.


Asunto(s)
Trasplante de Órganos , Trasplantes , Animales , Xenoinjertos , Humanos , Modelos Animales , Trasplante Heterólogo
11.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-34884740

RESUMEN

Extracellular vesicles (EV) derived from mesenchymal stromal cells (MSC) are a potential therapy for immunological and degenerative diseases. However, large-scale production of EV free from contamination by soluble proteins is a major challenge. The generation of particles from isolated membranes of MSC, membrane particles (MP), may be an alternative to EV. In the present study we generated MP from the membranes of lysed MSC after removal of the nuclei. The yield of MP per MSC was 1 × 105 times higher than EV derived from the same number of MSC. To compare the proteome of MP and EV, proteomic analysis of MP and EV was performed. MP contained over 20 times more proteins than EV. The proteins present in MP evidenced a multi-organelle origin of MP. The projected function of the proteins in EV and MP was very different. Whilst proteins in EV mainly play a role in extracellular matrix organization, proteins in MP were interconnected in diverse molecular pathways, including protein synthesis and degradation pathways and demonstrated enzymatic activity. Treatment of MSC with IFNγ led to a profound effect on the protein make up of EV and MP, demonstrating the possibility to modify the phenotype of EV and MP through modification of parent MSC. These results demonstrate that MP are an attractive alternative to EV for the development of potential therapies. Functional studies will have to demonstrate therapeutic efficacy of MP in preclinical disease models.


Asunto(s)
Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteoma , Membrana Celular/metabolismo , Humanos , Interferón gamma , Proteómica
12.
Transpl Int ; 33(8): 833-840, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32237237

RESUMEN

Regenerative medicine is emerging as a novel field in organ transplantation. In September 2019, the European Cell Therapy and Organ Regeneration Section (ECTORS) of the European Society for Organ Transplantation (ESOT) held its first meeting to discuss the state-of-the-art of regenerative medicine in organ transplantation. The present article highlights the key areas of interest and major advances in this multidisciplinary field in organ regeneration and discusses its implications for the future of organ transplantation.


Asunto(s)
Trasplante de Órganos , Medicina Regenerativa , Tratamiento Basado en Trasplante de Células y Tejidos , Regeneración
13.
Int J Mol Sci ; 21(22)2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-33238629

RESUMEN

Primary Sclerosing Cholangitis (PSC) is a progressive liver disease for which there is no effective medical therapy. PSC belongs to the family of immune-mediated biliary disorders and it is characterized by persistent biliary inflammation and fibrosis. Here, we explored the possibility of using extracellular vesicles (EVs) derived from human, bone marrow mesenchymal stromal cells (MSCs) to target liver inflammation and reduce fibrosis in a mouse model of PSC. Five-week-old male FVB.129P2-Abcb4tm1Bor mice were intraperitoneally injected with either 100 µL of EVs (± 9.1 × 109 particles/mL) or PBS, once a week, for three consecutive weeks. One week after the last injection, mice were sacrificed and liver and blood collected for flow cytometry analysis and transaminase quantification. In FVB.129P2-Abcb4tm1Bor mice, EV administration resulted in reduced serum levels of alkaline phosphatase (ALP), bile acid (BA), and alanine aminotransferase (ALT), as well as in decreased liver fibrosis. Mechanistically, we observed that EVs reduce liver accumulation of both granulocytes and T cells and dampen VCAM-1 expression. Further analysis revealed that the therapeutic effect of EVs is accompanied by the inhibition of NFkB activation in proximity of the portal triad. Our pre-clinical experiments suggest that EVs isolated from MSCs may represent an effective therapeutic strategy to treat patients suffering from PSC.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Colangitis Esclerosante/terapia , Inflamación/terapia , Hígado/metabolismo , Alanina Transaminasa/sangre , Fosfatasa Alcalina/sangre , Animales , Ácidos y Sales Biliares/sangre , Colangitis Esclerosante/sangre , Colangitis Esclerosante/genética , Colangitis Esclerosante/patología , Modelos Animales de Enfermedad , Vesículas Extracelulares/genética , Regulación de la Expresión Génica/efectos de los fármacos , Granulocitos/patología , Humanos , Inflamación/sangre , Inflamación/genética , Inflamación/patología , Hígado/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Noqueados , Linfocitos T/patología , Molécula 1 de Adhesión Celular Vascular/genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
14.
Stem Cells ; 36(4): 602-615, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29341339

RESUMEN

Mesenchymal stem or stromal cells (MSC) are under investigation as a potential immunotherapy. MSC are usually administered via intravenous infusion, after which they are trapped in the lungs and die and disappear within a day. The fate of MSC after their disappearance from the lungs is unknown and it is unclear how MSC realize their immunomodulatory effects in their short lifespan. We examined immunological mechanisms determining the fate of infused MSC and the immunomodulatory response associated with it. Tracking viable and dead human umbilical cord MSC (ucMSC) in mice using Qtracker beads (contained in viable cells) and Hoechst33342 (staining all cells) revealed that viable ucMSC were present in the lungs immediately after infusion. Twenty-four hours later, the majority of ucMSC were dead and found in the lungs and liver where they were contained in monocytic cells of predominantly non-classical Ly6Clow phenotype. Monocytes containing ucMSC were also detected systemically. In vitro experiments confirmed that human CD14++ /CD16- classical monocytes polarized toward a non-classical CD14++ CD16+ CD206+ phenotype after phagocytosis of ucMSC and expressed programmed death ligand-1 and IL-10, while TNF-α was reduced. ucMSC-primed monocytes induced Foxp3+ regulatory T cell formation in mixed lymphocyte reactions. These results demonstrate that infused MSC are rapidly phagocytosed by monocytes, which subsequently migrate from the lungs to other body sites. Phagocytosis of ucMSC induces phenotypical and functional changes in monocytes, which subsequently modulate cells of the adaptive immune system. It can be concluded that monocytes play a crucial role in mediating, distributing, and transferring the immunomodulatory effect of MSC. Stem Cells 2018;36:602-615.


Asunto(s)
Inmunomodulación , Pulmón/inmunología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Fagocitosis , Animales , Antígeno B7-H1/inmunología , Xenoinjertos , Humanos , Interleucina-10/inmunología , Masculino , Ratones , Factor de Necrosis Tumoral alfa/inmunología
15.
Int J Mol Sci ; 20(14)2019 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-31340593

RESUMEN

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.


Asunto(s)
Rastreo Celular/métodos , Glomérulos Renales/ultraestructura , Trasplante de Riñón , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Perfusión/métodos , Adipocitos/citología , Adipocitos/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Diferenciación Celular , Colorantes Fluorescentes/metabolismo , Humanos , Glomérulos Renales/cirugía , Trasplante de Células Madre Mesenquimatosas/instrumentación , Células Madre Mesenquimatosas/fisiología , Microscopía Fluorescente , Preservación de Órganos/métodos , Compuestos Orgánicos/metabolismo , Perfusión/instrumentación , Porcinos , Temperatura , Trasplante Heterólogo
16.
Cytotherapy ; 20(7): 919-929, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29934259

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) are studied for their immunotherapeutic potential. Prior to therapeutic use, MSCs are culture expanded to obtain the required cell numbers and, to improve their efficacy, MSCs may be primed in vitro. Culture expansion and priming induce phenotypical and functional changes in MSCs and thus standardisation and quality control measurements come in need. We investigated the impact of priming and culturing on MSC DNA methylation and examined the use of epigenetic profiling as a quality control tool. METHODS: Human umbilical cord-derived MSCs (ucMSCs) were cultured for 3 days with interferon (IFN)γ, transforming growth factor (TGF)ß or a multi-factor combination (MC; IFNγ, TGFß and retinoic acid). In addition, ucMSCs were culture expanded for 14 days. Phenotypical changes and T-cell proliferation inhibition capacity were examined. Genome-wide DNA methylation was measured with Infinium MethylationEPIC Beadchip. RESULTS: Upon priming, ucMSCs exhibited a different immunophenotype and ucMSC(IFNγ) and ucMSC(MC) had an increased capacity to inhibit T-cell proliferation. DNA methylation patterns were minimally affected by priming, with only one significantly differentially methylated site (DMS) in IFNγ- and MC-primed ucMSCs associated with autophagy activity. In contrast, 14 days after culture expansion, ucMSCs displayed minor phenotypical and functional changes but showed >4000 significantly DMSs, mostly concerning genes involved in membrane composition, cell adhesion and transmembrane signalling. DISCUSSION: These data show that DNA methylation of MSCs is only marginally affected by priming, whereas culture expansion and subsequent increased cellular interactions have a large impact on methylation. On account of this study, we suggest that DNA methylation analysis is a useful quality control tool for culture expanded therapeutic MSCs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Epigénesis Genética , Células Madre Mesenquimatosas/metabolismo , Cordón Umbilical/citología , Biomarcadores/metabolismo , Forma de la Célula , Células Cultivadas , Metilación de ADN/genética , Humanos , Inmunofenotipificación , Interferón gamma/metabolismo , Células Madre Mesenquimatosas/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo
18.
Stem Cells ; 34(2): 483-92, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26661518

RESUMEN

The role of interleukin 1 receptor antagonist (IL1RA) in mediating the immunosuppressive effect of mesenchymal stem/stromal cells (MSCs) has been reported in several studies. However, how MSC-derived IL1RA influences the host response has not been clearly investigated. We therefore derived MSCs from the bone marrow of IL1RA knockout mice and evaluated their immunosuppressive effect on different immune cell subsets. IL1RA deficient (IL1RA(-/-) ) or wild type (wt) MSCs inhibited to the same extend the proliferation of T lymphocytes. On the contrary, IL1RA(-/-) MSCs were less effective than wt MSCs to induce in vitro the macrophage polarization from M1 to M2 phenotype secreting IL10 and exerting a suppressive effect on CD4(+) T cells. Moreover compared with wt MSCs, IL1RA(-/-) MSCs did not efficiently support the survival of quiescent B lymphocytes and block their differentiation toward CD19(+) CD138(+) plasmablasts secreting IgG antibodies. The effectiveness of IL1RA secreted by MSCs in controlling inflammation was further shown in vivo using the collagen-induced arthritis murine model. MSCs lacking IL1RA expression were unable to protect mice from arthritic progression and even worsened clinical signs, as shown by higher arthritic score and incidence than control arthritic mice. IL1RA(-/-) MSCs were not able to decrease the percentage of Th17 lymphocytes and increase the percentage of Treg cells as well as decreasing the differentiation of B cells toward plasmablasts. Altogether, our results provide evidence of the key role of IL1RA secreted by MSCs to both control the polarization of macrophages toward a M2 phenotype and inhibit B cell differentiation in vivo.


Asunto(s)
Linfocitos B/metabolismo , Diferenciación Celular , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Linfocitos B/inmunología , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Macrófagos/inmunología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Noqueados , Células Th17/inmunología , Células Th17/metabolismo
19.
Cytotherapy ; 19(7): 798-807, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28462821

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) are used as experimental immunotherapy. Extensive culture expansion is necessary to obtain clinically relevant cell numbers, although the impact on MSCs stability and function is unclear. This study investigated the effects of long-term in vitro expansion on the stability and function of MSCs. METHODS: Human bone marrow-derived (bmMSCs) and umbilical cord-derived (ucMSCs) MSCs were in vitro expanded. During expansion, their proliferative capacity was examined. At passages 4, 8 and 12, analyses were performed to investigate the ploidy, metabolic stability, telomere length and immunophenotype. In addition, their potential to suppress lymphocyte proliferation and susceptibility to natural killer cell lysis was examined. RESULTS: BmMSCs and ucMSCs showed decreasing proliferative capacity over time, while their telomere lengths and mitochondrial activity remained stable. Percentage of aneuploidy in cultures was unchanged after expansion. Furthermore, expression of MSC markers and markers associated with stress or aging remained unchanged. Reduced capacity to suppress CD4 and CD8 T-cell proliferation was observed for passage 8 and 12 bmMSCs and ucMSCs. Finally, susceptibility of bmMSCs and ucMSCs to NK-cell lysis remained stable. CONCLUSIONS: We showed that after long-term expansion, phenotype of bmMSCs and ucMSCs remains stable and cells exhibit similar immunogenic properties compared with lower passage cells. However, immunosuppressive properties of MSCs are reduced. These findings reveal the consequences of application of higher passage MSCs in the clinic, which will help increase the yield of therapeutic MSCs but may interfere with their efficacy.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Mesenquimatosas/fisiología , Cordón Umbilical/citología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/fisiología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Inmunofenotipificación , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/fisiología , Células Madre Mesenquimatosas/citología , Ploidias , Embarazo , Homeostasis del Telómero , Factores de Tiempo
20.
Cytotherapy ; 18(8): 957-969, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27288309

RESUMEN

BACKGROUND AIMS: In regenerative medicine, the use of allogeneic cells could enable the development of "off the shelf" therapies for patients with critical size bone defects, reducing limitations observed with the use of autologous cells, such as cost and time to treat the patient. The idea of the use of allogeneic bone marrow mesenchymal stromal cells (BMSCs) has been of interest in tissue engineering studies. However, little is known about the properties of these cells upon differentiation. Chondrogenically differentiated BMSCs have already been shown to form endochondral bone in immunodeficient and immunocompetent animals. The success of this bone formation is dependent on the host's endogenous cells. This study investigates the interactions between allogeneic chondrogenically differentiated human bone marrow mesenchymal stromal cell (hBMSC) pellets and T lymphocytes in vitro. METHODS: Non-chondrogenic (-transforming growth factor (TGF)ß3) and chondrogenic hBMSC (+TGFß3) pellets were directly co-cultured with unstimulated and CD3/CD28-stimulated peripheral blood mononuclear cells (PBMCs) for 7 days. hBMSC pellets from the co-culture were either fixed for histological analysis or quantitative real time polymerase chain reaction (qRT-PCR). PBMCs were harvested for flow cytometry. RESULTS: Flow cytometic analysis revealed that chondrogenically differentiated hBMSC pellets did not alter the number or proliferation of CD4+, CD8+ T cells or FoxP3+ T regulatory cells (CD4+CD25+CD127-). Chondrogenic hBMSC pellets did not induce immunogenic responses in unstimulated PBMCs. Infiltrating CD3 T cells were found in the matrix of hBMSC pellets. Furthermore, qRT-PCR demonstrated low levels of T-cell activation genes (CD25, CD69, PRF1 and GZMB) in addition to low gene expression levels of the pro-inflammatory gene tumor necrosis factor alpha (TNFα) in chondrogenically differentiated hBMSC pellets cultured with unstimulated PBMCs in comparison with non-chondrogenic hBMSC pellets. CONCLUSIONS: Collectively the results of this study demonstrate that allogeneic chondrogenically differentiated hBMSC pellets are non-immunogenic and do not induce the activation of destructive T-cell responses in vitro.


Asunto(s)
Diferenciación Celular , Condrocitos/fisiología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/fisiología , Linfocitos T/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/fisiología , Células Cultivadas , Condrogénesis/fisiología , Técnicas de Cocultivo , Humanos , Inmunomodulación/fisiología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/fisiología , Activación de Linfocitos , Masculino , Células Madre Mesenquimatosas/citología , Osteogénesis , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA