Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
8.
Transpl Int ; 26(6): 651-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23682671

RESUMEN

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


Asunto(s)
Suero Antilinfocítico/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Animales , Suero Antilinfocítico/metabolismo , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunosupresores/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Conejos
9.
Kidney Int ; 82(7): 748-58, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22695328

RESUMEN

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


Asunto(s)
Tejido Adiposo/metabolismo , Enfermedades Renales/metabolismo , Células Madre Mesenquimatosas/metabolismo , 5'-Nucleotidasa/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Adulto , Anciano , Antígenos CD/metabolismo , Apoptosis , Biomarcadores/metabolismo , Antígenos CD28/metabolismo , Complejo CD3/metabolismo , Estudios de Casos y Controles , Moléculas de Adhesión Celular Neuronal/metabolismo , Diferenciación Celular , Proliferación Celular , Separación Celular/métodos , Forma de la Célula , Células Cultivadas , Técnicas de Cocultivo , Endoglina , Femenino , Proteínas Fetales/metabolismo , Citometría de Flujo , Proteínas Ligadas a GPI/metabolismo , Regulación de la Expresión Génica , Humanos , Inmunofenotipificación/métodos , Mediadores de Inflamación/metabolismo , Enfermedades Renales/sangre , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Antígenos Comunes de Leucocito/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores de Superficie Celular/metabolismo , Factores de Tiempo , Trasplante Autólogo , Uremia/sangre , Uremia/inmunología
10.
Front Immunol ; 12: 650522, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897698

RESUMEN

Proinflammatory stimuli lead to endothelial injury, which results in pathologies such as cardiovascular diseases, autoimmune diseases, and contributes to alloimmune responses after organ transplantation. Both mesenchymal stromal cells (MSC) and the extracellular vesicles (EV) released by them are widely studied as regenerative therapy for the endothelium. However, for therapeutic application, the manipulation of living MSC and large-scale production of EV are major challenges. Membrane particles (MP) generated from MSC may be an alternative to the use of whole MSC or EV. MP are nanovesicles artificially generated from the membranes of MSC and possess some of the therapeutic properties of MSC. In the present study we investigated whether MP conserve the beneficial MSC effects on endothelial cell repair processes under inflammatory conditions. MP were generated by hypotonic shock and extrusion of MSC membranes. The average size of MP was 120 nm, and they showed a spherical shape. The effects of two ratios of MP (50,000; 100,000 MP per target cell) on human umbilical vein endothelial cells (HUVEC) were tested in a model of inflammation induced by TNFα. Confocal microscopy and flow cytometry showed that within 24 hours >90% of HUVEC had taken up MP. Moreover, MP ended up in the lysosomes of the HUVEC. In a co-culture system of monocytes and TNFα activated HUVEC, MP did not affect monocyte adherence to HUVEC, but reduced the transmigration of monocytes across the endothelial layer from 138 ± 61 monocytes per microscopic field in TNFα activated HUVEC to 61 ± 45 monocytes. TNFα stimulation induced a 2-fold increase in the permeability of the HUVEC monolayer measured by the translocation of FITC-dextran to the lower compartment of a transwell system. At a dose of 1:100,000 MP significantly decreased endothelial permeability (1.5-fold) respect to TNFα Stimulated HUVEC. Finally, MP enhanced the angiogenic potential of HUVEC in an in vitro Matrigel assay by stimulating the formation of angiogenic structures, such as percentage of covered area, total tube length, total branching points, total loops. In conclusion, MP show regenerative effects on endothelial cells, opening a new avenue for treatment of vascular diseases where inflammatory processes damage the endothelium.


Asunto(s)
Tejido Adiposo/citología , Vesículas Extracelulares/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Adhesión Celular/inmunología , Permeabilidad de la Membrana Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Microscopía por Crioelectrón , ADN/genética , ADN/aislamiento & purificación , Vesículas Extracelulares/genética , Vesículas Extracelulares/ultraestructura , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Microscopía Electrónica de Transmisión , Monocitos/citología , Tamaño de la Partícula , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
PLoS One ; 16(3): e0248415, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33730089

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with limited treatment options in which the telomere shortening is a strong predictive factor of poor prognosis. Mesenchymal stromal cells (MSC) administration is probed in several experimental induced lung pathologies; however, MSC might stimulate fibrotic processes. A therapy that avoids MSC side effects of transformation would be an alternative to the use of living cells. Membranes particles (MP) are nanovesicles artificially generated from the membranes of MSC containing active enzymes involved in ECM regeneration. We aimed to investigate the anti-fibrotic role of MP derived from MSC in an in vitro model of pulmonary fibrosis. METHODS: Epithelial cells (A549) and lung fibroblasts, from IPF patients with different telomere length, were co-cultured with MP and TGF-ß for 48h and gene expression of major pro-fibrotic markers were analyzed. RESULTS: About 90% of both types of cells effectively took up MP without cytotoxic effects. MP decreased the expression of profibrotic proteins such as Col1A1, Fibronectin and PAI-1, in A549 cells. In fibroblasts culture, there was a different response in the inhibitory effect of MP on some pro-fibrotic markers when comparing fibroblast from normal telomere length patients (FN) versus short telomere length (FS), but both types showed an inhibition of Col1A1, Tenascin-c, PAI-1 and MMP-1 gene expression after MP treatment. CONCLUSIONS: MP conserve some of the properties attributed to the living MSC. This study shows that MP target lung cells, via which they may have a broad anti-fibrotic effect.


Asunto(s)
Micropartículas Derivadas de Células/trasplante , Fibrosis Pulmonar Idiopática/terapia , Células Madre Mesenquimatosas/citología , Nanopartículas/uso terapéutico , Cultivo Primario de Células/métodos , Células A549 , Adulto , Anciano , Técnicas de Cocultivo , Femenino , Fibroblastos , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/citología , Pulmón/patología , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Persona de Mediana Edad , Grasa Subcutánea/citología , Acortamiento del Telómero
12.
Front Immunol ; 12: 651109, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790914

RESUMEN

Mesenchymal stromal cells (MSC) are a promising therapy for inflammatory diseases. However, MSC are large and become trapped in the lungs after intravenous infusion, where they have a short survival time. To steer MSC immunoregulatory therapy beyond the lungs, we generated nm-sized particles from MSC membranes (membrane particles, MP), which have immunomodulatory properties, and investigated their internalization and mode of interaction in macrophages subtypes and human umbilical vein endothelial cells (HUVEC) under control and inflammatory conditions. We found that macrophages and HUVEC take up MP in a dose, time, and temperature-dependent manner. Specific inhibitors for endocytotic pathways revealed that MP internalization depends on heparan sulfate proteoglycan-, dynamin-, and clathrin-mediated endocytosis but does not involve caveolin-mediated endocytosis. MP uptake also involved the actin cytoskeleton and phosphoinositide 3-kinase, which are implicated in macropinocytosis and phagocytosis. Anti-inflammatory M2 macrophages take up more MP than pro-inflammatory M1 macrophages. In contrast, inflammatory conditions did not affect the MP uptake by HUVEC. Moreover, MP induced both anti- and pro-inflammatory responses in macrophages and HUVEC by affecting gene expression and cell surface proteins. Our findings on the mechanisms of uptake of MP under different conditions help the development of target-cell specific MP therapy to modulate immune responses.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Micropartículas Derivadas de Células/inmunología , Células Madre Mesenquimatosas/citología , Micropartículas Derivadas de Células/trasplante , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Voluntarios Sanos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Macrófagos/inmunología , Células Madre Mesenquimatosas/inmunología , Fagocitosis/inmunología , Pinocitosis/inmunología , Cultivo Primario de Células , Grasa Subcutánea/citología
13.
Stem Cells Dev ; 30(22): 1103-1114, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34549597

RESUMEN

Human-induced pluripotent stem cell (iPSC)-derived kidney organoids have the potential to advance studies to kidney development and disease. However, reproducible generation of kidney organoids is a challenge. A large variability in the percentage of nephron structures and the expression of kidney-specific genes was observed among organoids, showing no association with iPSC lines. To associate the quality of kidney organoid differentiation with predictive markers, a ranking system was developed based on the ratio of nephron structure determined by histological examination. Well-differentiated organoids were defined as organoids with >30% nephron structure and vice versa. Subsequently, correlations were made with expression profiles of iPSC markers, early kidney development markers, and fibrosis markers. Higher expression of sex-determining region Y-box 2 (SOX2) during differentiation was associated with poorly differentiated kidney organoid. Furthermore, early secretion of basic fibroblast growth factor (FGF2) predicted poorly differentiated kidney organoid. Of interest, whereas cadherin-1 (CDH1) expression in kidney organoids indicates distal tubules formation, onefold higher CDH1 expression in iPSC predicted poor differentiation. High expression of the stromal progenitor marker Forkhead Box D1 (FOXD1) and significantly increased TGFß levels were found in well-differentiated kidney organoids. These early expression profiles could predict the outcome of kidney organoid formation. This study helps to improve the robustness of kidney organoid protocols.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Diferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Humanos , Riñón , Organoides
14.
Stem Cells Dev ; 29(1): 38-48, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31696786

RESUMEN

Mesenchymal stromal cells (MSCs) are attractive candidates for immunomodulatory cell therapy. However, it remains unknown how far therapeutic efficacy and potency are dependent on the dosage and activity of the MSCs. We previously observed that infusion of MSCs leads to rapid and transient changes in cytokine expression in blood, lung, and liver. In the present study, increasing doses of syngeneic adipose tissue-derived MSCs were infused in healthy mice and systemic changes in G-CSF, IL6, IL-10, and CXCL5 were detected 2 h after administration of 3 × 105 MSCs per animal, but not at lower doses. In lung and liver tissue, dose-dependent effects of MSCs on cytokine mRNA expression levels were detected from doses as low as 3 × 103 MSCs. Infusion of secretome-deficient or IFNγ-activated MSCs in healthy mice had similar effects on systemic cytokine levels as control MSCs, suggesting that in vivo at least the initial systemic effect of MSC administration is independent of the level of activity of MSCs, but depends on the response of host cells to MSCs. The results of this study reveal a rapid dose-dependent effect of MSCs and stress the important role of host cells in MSC treatment. This knowledge contributes to the design of rational MSC trials and to the quest for clinical efficacy of MSC therapy.


Asunto(s)
Tejido Adiposo/citología , Inmunomodulación , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Recuento de Células , Células Cultivadas , Citocinas/sangre , Citocinas/genética , Citocinas/metabolismo , Femenino , Expresión Génica , Humanos , Interferones/farmacología , Hígado/metabolismo , Pulmón/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Factores de Tiempo
15.
Transpl Immunol ; 18(3): 250-4, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18047933

RESUMEN

Previously, we demonstrated in heart transplant patients that FOXP3, a gene required for the development and function of regulatory T cells, was highly expressed in the graft during an acute cellular rejection. In this study, we analyzed whether the FOXP3 gene expression in the peripheral blood also reflects anti-donor immune responses, and therefore may provide clues for non-invasive detection of non-responsiveness or acute rejection. We examined the FOXP3 expression patterns of peripheral blood mononuclear cells (PBMC; n=69) of 19 heart transplant patients during quiescence and rejection in comparison with those of endomyocardial biopsies (EMB; n=75) of 24 heart transplant patients. While the FOXP3 mRNA levels were abundantly expressed in rejecting EMB (ISHLT rejection grade>1R) compared with EMB without histological evidence of myocardial damage (ISHLT rejection grade 0R-1R; p=0.003), no association with rejection or non-responsiveness was found for the FOXP3 mRNA levels in the peripheral blood. Thus, in contrast to intragraft FOXP3 gene expression, the peripheral FOXP3 mRNA levels lack correlation with anti-donor immune responses in the graft, and, consequently, FOXP3 does not appear to be a potential candidate gene for non-invasive diagnosis of non-responsiveness or rejection.


Asunto(s)
Factores de Transcripción Forkhead/genética , Rechazo de Injerto , Trasplante de Corazón/inmunología , Leucocitos Mononucleares/metabolismo , Adulto , Anciano , Biopsia , Femenino , Factores de Transcripción Forkhead/sangre , Expresión Génica , Supervivencia de Injerto/inmunología , Humanos , Masculino , Persona de Mediana Edad , Miocardio/patología , ARN Mensajero/sangre , ARN Mensajero/genética
16.
Transplantation ; 83(11): 1477-84, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17565321

RESUMEN

BACKGROUND: Regulatory FOXP3+ T cells control immune responses of effector T cells. However, whether these cells regulate antidonor responses in the graft of cardiac allograft patients is unknown. Therefore, we analyzed the gene expression profiles of regulatory and effector T-cell markers during immunological quiescence and acute rejection. METHODS: Quantitative real-time polymerase chain reaction was used to analyze mRNA expression levels in time-zero specimens (n=24) and endomyocardial biopsies (EMB; n=72) of cardiac allograft patients who remained free from rejection (nonrejectors; n=12) and patients with at least one histologically proven acute rejection episode (rejectors; International Society for Heart and Lung Transplantation [ISHLT] rejection grade>2; n=12). RESULTS: For all analyzed regulatory and effector T-cell markers, mRNA expression levels were increased in biopsies taken after heart transplantation compared with those in time-zero specimens. Posttransplantation, the FOXP3 mRNA levels were higher in EMB assigned to a higher ISHLT rejection grade than the biopsies with grade 0: the highest mRNA levels were detected in the rejection biopsies (rejection grade>2; P=0.003). In addition, the mRNA levels of CD25, glucocorticoid-induced TNF receptor family-related gene, cytotoxic T lymphocyte-associated antigen 4, interleukin-2, and granzyme B were also significantly higher in rejecting EMB than in nonrejecting EMB (rejection grade

Asunto(s)
Factores de Transcripción Forkhead/genética , Trasplante de Corazón/inmunología , Miocardio/metabolismo , ARN Mensajero/metabolismo , Enfermedad Aguda , Adolescente , Adulto , Anciano , Antígenos CD/genética , Antígenos de Diferenciación/genética , Antígeno CTLA-4 , Endocardio/metabolismo , Femenino , Expresión Génica , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Rechazo de Injerto/genética , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Granzimas/genética , Humanos , Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/genética , Masculino , Persona de Mediana Edad , Periodo Posoperatorio , Índice de Severidad de la Enfermedad , Donantes de Tejidos , Trasplante Homólogo
17.
Transplantation ; 83(11): 1485-92, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17565322

RESUMEN

BACKGROUND: Interleukin (IL)-21 is the most recently described cytokine that signals via the common cytokine receptor (gammac), is produced by activated CD4+ T-cells, and regulates expansion and effector function of CD8+ T-cells. MATERIALS: To explore the actions of IL-21 with other gammac-dependent cytokines in alloreactivity, mRNA expression of IL-21, IL-21R alpha-chain, and IL-2 proliferation and cytotoxicity was measured after stimulation in mixed lymphocyte reactions. Additionally, IL-21 and IL-21R alpha-chain expression was studied in biopsies of heart transplant patients. RESULTS: Analysis of mRNA expression levels of allostimulated T-cells showed a 10-fold induction of IL-21 and IL-21R alpha-chain. Interestingly, induction of IL-21 was highly dependent on IL-2 (as in the presence of anti-IL-2, anti-IL-2R alpha-chain, and the immunosuppressive drugs cyclosporine A, tacrolimus, and rapamycin) the transcription of IL-21 was almost completely inhibited, whereas in the presence of exogenous IL-2 the mRNA expression of IL-21 was even more upregulated. IL-21 functioned as a costimulator for IL-2 to augment proliferation and cytotoxic responses, while blockade of the IL-2 route abrogated these functions of IL-21. Blockade of the IL-21 route by anti-IL-21R alpha-chain monoclonal antibodies inhibited the proliferation of alloactivated T-cells. Also, in vivo alloreactivity was associated with IL-21/IL-21R alpha-chain expression. After heart transplantation, the highest intragraft IL-21, IL-21R alpha-chain, and IL-2 mRNA expression levels were measured during acute rejection (P<0.001, P=0.01, P=0.03). CONCLUSION: IL-21 is a critical cytokine for IL-2 dependent immune processes. Blockade of the IL-21 pathway may provide a new perspective for the treatment of allogeneic responses in patients after transplantation.


Asunto(s)
Rechazo de Injerto/fisiopatología , Trasplante de Corazón , Interleucina-2/metabolismo , Interleucinas/metabolismo , Proliferación Celular , Estudios de Cohortes , Citotoxicidad Inmunológica , Endocardio/metabolismo , Rechazo de Injerto/sangre , Rechazo de Injerto/genética , Rechazo de Injerto/metabolismo , Humanos , Terapia de Inmunosupresión , Interleucinas/genética , Prueba de Cultivo Mixto de Linfocitos , Miocardio/metabolismo , Periodo Posoperatorio , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores de Interleucina-21/genética , Receptores de Interleucina-21/metabolismo , Linfocitos T/patología
18.
Front Immunol ; 8: 1042, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28894451

RESUMEN

The immunomodulatory capacity of mesenchymal stem or stromal cells (MSC) makes them a promising tool for treatment of immune disease and organ transplantation. The effects of MSC on B cells are characterized by an abrogation of plasmablast formation and induction of regulatory B cells (Bregs). It is, however, unknown how MSC interact with B cells under inflammatory conditions. In this study, adipose tissue-derived MSC were pretreated with 50 ng/ml IFN-γ for 96 h (MSC-IFN-γ) to simulate inflammatory conditions. Mature B cells were obtained from spleens by CD43- selection. B cells were co-cultured with MSC and stimulated with anti-IgM, anti-CD40, and IL-2; and after 7 days, B cell proliferation, phenotype, Immunoglobulin-G (IgG), and IL-10 production were analyzed. MSC did not inhibit B cell proliferation but increased the percentage of CD38high CD24high B cells (Bregs) and IL-10 production, while MSC-IFN-γ significantly reduced B cell proliferation and inhibited IgG production by B cells in a more potent fashion but did not induce Bregs or IL-10 production. Both MSC and MSC-IFN-γ required proximity to target cells and being metabolically active to exert their effects. Indoleamine 2,3 dioxygenase expression was highly induced in MSC-IFN-γ and was responsible of the anti-proliferative and Breg reduction since addition of tryptophan (TRP) restored MSC properties. Immunological conditions dictate the effect of MSC on B cell function. Under immunological quiescent conditions, MSC stimulate Breg induction; whereas, under inflammatory conditions, MSC inhibit B cell proliferation and maturation through depletion of TRP. This knowledge is useful for customizing MSC therapy for specific purposes by appropriate pretreatment of MSC.

19.
Sci Rep ; 7(1): 12100, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28935974

RESUMEN

Mesenchymal stromal cells (MSC) are a promising therapy for immunological disorders. However, culture expanded MSC are large and get trapped in the capillary networks of the lungs after intravenous infusion, where they have a short survival time. Hypothetically, living cells are a risk for tumor formation. To reduce risks associated with MSC infusion and improve the distribution in the body, we generated membrane particles (MP) of MSC and MSC stimulated with IFN-γ (MPγ). Tracking analysis and electron microscopy indicated that the average size of MP was 120 nm, and they showed a round shape. MP exhibited ATPase, nucleotidase and esterase activity, indicating they are enzymatically active. MP and MPγ did not physically interact with T cells and had no effect on CD4+ and CD8+ T cells proliferation. However, MP and MPγ selectively bound to monocytes and decreased the frequency of pro-inflammatory CD14+CD16+ monocytes by induction of selective apoptosis. MP and MPγ increased the percentage of CD90 positive monocytes, and MPγ but not MP increased the percentage of anti-inflammatory PD-L1 monocytes. MPγ increased mRNA expression of PD-L1 in monocytes. These data demonstrate that MP have immunomodulatory properties and have potential as a novel cell-free therapy for treatment of immunological disorders.


Asunto(s)
Antígeno B7-H1/inmunología , Micropartículas Derivadas de Células/inmunología , Mediadores de Inflamación/inmunología , Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Tejido Adiposo/citología , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Micropartículas Derivadas de Células/ultraestructura , Células Cultivadas , Expresión Génica/efectos de los fármacos , Inmunomodulación/inmunología , Mediadores de Inflamación/metabolismo , Interferón gamma/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Células Madre Mesenquimatosas/metabolismo , Microscopía Electrónica de Transmisión , Monocitos/metabolismo , Tamaño de la Partícula , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
20.
Stem Cells Int ; 2017: 4960831, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28642794

RESUMEN

Mesenchymal stem cells derived from adipose tissue (ASC) have immune regulatory function, which makes them interesting candidates for cellular therapy. ASC cultures are however heterogeneous in phenotype. It is unclear whether all ASC contribute equally to immunomodulatory processes. ASC are also responsive to cytokine stimulation, which may affect the ratio between more and less potent ASC populations. In the present study, we determined IL-6 receptor (CD126 and CD130 subunits) and IFN-γ receptor (CD119) expression on ASC by flow cytometry. The production of IL-6 and IFN-γ was measured by ELISA and the frequency of IL-6 and IFN-γ secreting cells by ELISPOT. The results showed that ASC did not express CD126, and only 10-20% of ASC expressed CD130 on their surface, whereas 18-31% of ASC expressed CD119. ASC produced high levels of IL-6 and 100% of ASC were capable of secreting IL-6. Stimulation by IFN-γ or TGF-ß had no effect on IL-6 secretion by ASC. IFN-γ was produced by only 1.4% of ASC, and TGF-ß significantly increased the frequency to 2.7%. These results demonstrate that ASC cultures are heterogeneous in their cytokine secretion and receptor expression profiles. This knowledge can be employed for selection of potent, cytokine-producing, or responsive ASC subsets for cellular immunotherapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA