Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Exp Immunol ; 208(3): 255-267, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35439818

RESUMEN

FOXP3+ regulatory T cells (Tregs) are central to maintaining peripheral tolerance and immune homeostasis. They have the potential to be developed as a cellular therapy to treat various clinical ailments such as autoimmune disorders, inflammatory diseases and to improve transplantation outcomes. However, a major question remains whether Tregs can persist and exert their function effectively in a disease state, where a broad spectrum of inflammatory mediators could inactivate Tregs. In this study, we investigated the potential of mesenchymal stem cell (MSC)-derived exosomes to promote and sustain Tregs function. MSC-conditioned media (MSC-CM) cultured Tregs were more suppressive in both polyclonal and allogeneic responses and were resistant to inflammatory stimulation in vitro compared with the controls. A similar enhancement of Treg function was also observed by culturing Tregs with MSC-derived exosomes alone. The enhanced suppressive activity and stability of Treg cultured in MSC-CM was reduced when exosomes were depleted from MSC-CM. We identified that MSC-derived exosomes could upregulate the expression of LC3(II/I), phosphorylate Jak3 and Stat5 to promote Treg survival, and regulate FOXP3 expression in Tregs. Overall, our study demonstrates that MSC-derived exosomes are capable of enhancing Hucb-Tregs function and stability by activating autophagy and Stat5 signalling pathways. Our findings provide a strong rationale for utilizing MSC-derived exosomes as an effective strategy to enhance Treg function, and improve the overall Tregs-based cell therapy landscape.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Exosomas/metabolismo , Sangre Fetal , Factores de Transcripción Forkhead/metabolismo , Humanos , Factor de Transcripción STAT5/metabolismo , Linfocitos T Reguladores
2.
Biochem Biophys Res Commun ; 495(4): 2512-2518, 2018 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-29278702

RESUMEN

Hypoxia and islet inflammation are involved in ß-cell failure in type 2 diabetes (T2D). Elevated plasma LPS levels have been verified in patients with T2D, and hypoxia occurs in islets of diabetic mice. Activation of inflammasomes in ischemic or hypoxic conditions was identified in various tissues. Here, we investigated whether hypoxia activates the inflammasome in ß cells and the possible mechanisms involved. In mouse insulinoma cell line 6 (MIN6), hypoxia (1% O2) primes the NLRP3 inflammasome along with NF-κB signaling activation. Our results demonstrate that hypoxia can activate the NLRP3 inflammasome in LPS-primed MIN6 to result in initiating the ß cell inflammatory response and cell death in vitro. Reactive oxygen species (ROS) and the thioredoxin-interacting protein (TXNIP) are up-regulated in response to hypoxia. Finally, the role of the ROS-TXNIP axis in mediating the activation of the NLRP3 inflammasome and cell death was characterized by pretreating with the ROS scavenger N-acetylcysteine (NAC) and performing TXNIP knockdown experiments in MIN6. Our data indicate for the first time that the inflammasome is involved in the inflammatory response and cell death in hypoxia-induced ß cells through the ROS-TXNIP-NLRP3 axis in vitro. This provides new insight into the relationship between hypoxia and inflammation in T2D.


Asunto(s)
Apoptosis/inmunología , Proteínas Portadoras/inmunología , Hipoxia de la Célula/inmunología , Inflamasomas/inmunología , Células Secretoras de Insulina/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Especies Reactivas de Oxígeno/inmunología , Tiorredoxinas/inmunología , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Inflamasomas/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Lipopolisacáridos , Ratones , FN-kappa B/inmunología
3.
Xenotransplantation ; 25(5): e12405, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29932262

RESUMEN

BACKGROUND: Hypoxia-induced damage is one of the key factors associated with islet graft dysfunction. Mesenchymal stem cells (MSCs) could be used to enhance the therapeutic effect of islet transplantation due to their paracrine potential such as exosomes. In this study, we investigated whether exosomes from human umbilical cord-derived MSC-conditioned medium (hu-MSC-CM) could increase the survival and function of neonatal porcine islet cell clusters (NICCs) exposed to hypoxia. METHODS: Neonatal porcine islet cell clusters were cultured with hu-MSC-CM, with or without exosomes, and native medium RPMI-1640 (Control) under hypoxic conditions (1% O2 ). The effects of exosomes on NICCs viability and function in vitro were examined by FACS, the Loops system, and the Extracellular Flux assay, respectively. RESULTS: Compared with NICCs cultured in RPMI-1640 medium and hu-MSC-CM without exosomes, the survival ratio, viability, and function increased in NICCs cultured in hu-MSC-CM with exosomes. CONCLUSIONS: This study found that hu-MSC-CM could protect NICCs from hypoxia-induced dysfunction, and exosomes played an important role in hypoxic resistance, suggesting a potential strategy to improve islet transplantation outcomes.


Asunto(s)
Exosomas/inmunología , Hipoxia , Islotes Pancreáticos/citología , Células Madre Mesenquimatosas/citología , Animales , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Humanos , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Porcinos , Trasplante Heterólogo/métodos
4.
Xenotransplantation ; 25(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29131417

RESUMEN

BACKGROUND: Neonatal pig islet-like cell clusters (NICC) are an attractive source of insulin-producing tissue for potential transplantation treatment of type 1 diabetic patients. However, a considerable loss of NICC after their transplantation due to apoptosis resulted from islet isolation and instant blood-mediated inflammatory reaction remains to be overcome. METHODS: EGM2 medium depleted with hydrocortisone and supplemented with 50 mmol/L isobutylmethylxanthine, 10 mmol/L nicotinamide, and 10 mmol/L glucose was used to culture NICC at day 1, the day after isolation and changed every other day. NICC cultured with EGM2 or control Ham's F-10 medium were collected at day 7 of culture for the following assays. The viability of NICC was evaluated by AO/EB staining and FACS. Static assay and oxygen consumption rate analysis were performed to assess the function of NICC. Insulin and glucagon gene expression were measured by real-time PCR. Tubing loops model and TUNEL assay were performed to confirm the apoptosis-resistant ability of NICC cultured with modified EGM2 medium. Serum starvation and hypoxia treatment were used to test the tolerant capability of NICC in the microenvironment of hypoxia/nutrient deficiency in vitro. The molecules involved in apoptosis pathways in NICC were analyzed by Western blotting. RESULTS: Compared with Ham's F-10 medium, culturing NICC with EGM2 medium led to increased number and viability of NICC with higher stimulation index, upregulated gene expression of both insulin and glucagon, and enhanced mitochondria function. Furthermore, fewer modified EGM2 medium cultured NICC were found under apoptosis when evaluated in an in vitro tubing loop model of IBMIR. Moreover, EGM2 medium cultured NICC demonstrated much less apoptotic cells under either serum starvation or hypoxia condition than their Ham's F-10 medium cultured counterparts. The enhanced capability of EGM2 cultured NICC to resist apoptosis was associated with their elevated protein levels of anti-apoptotic Bcl-2 family member Mcl-1. CONCLUSION: Culturing NICC with EGM2 provides a simple and effective approach not only to increase NICC yield, viability, and maturation but also to enhance their resistance to apoptosis to preserve the initial graft mass for successful islet xenotransplantation.


Asunto(s)
Apoptosis/inmunología , Xenoinjertos/inmunología , Islotes Pancreáticos/citología , Trasplante Heterólogo , Animales , Animales Recién Nacidos , Células Cultivadas , Diabetes Mellitus/inmunología , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Trasplante Heterólogo/métodos
5.
Xenotransplantation ; 24(5)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28771838

RESUMEN

BACKGROUND: For xenotransplantation to truly succeed, we must develop immunomodulatory strategies to suppress the xenoimmune response but by minimizing immunosuppression over the long term. Regulatory macrophages (Mreg) have been shown to suppress polyclonal T-cell proliferation in vitro and prolong allograft survival in vivo. However, the question of whether they are capable of suppressing xenoimmune responses remains unknown. This study assessed the potential of human Mreg to be used as an effective immunomodulatory method in xenotransplantation. METHODS: CD14+ monocytes selected from human peripheral blood mononuclear cells (PBMC) were cultured with macrophage colony-stimulating factor (M-CSF) for 7 days with IFN-γ added at day 6 for Mreg induction. Mreg phenotyping was performed by flow cytometric analysis, and the in vitro suppressive function was assessed by mixed lymphocyte reaction (MLR) using irradiated pig PBMC as the xenogeneic stimulator cells, human PBMC as responder cells, and autologous Mreg as suppressor cells. To assess mRNA expression of Mreg functional molecules indoleamine-2,3-dioxygenase (IDO), IL-10, inducible nitric oxide synthase (iNOS) and TGF-ß were measured by real-time PCR. Supernatants were collected from the MLR cultures for IDO activity assay by high-performance liquid chromatography (HPLC). The effects of the IDO inhibitor 1-D/L-methyl-tryptophan (1-MT), iNOS inhibitor NG -monomethyl-l-arginine (L-NMMA), and anti-IFN-γ or anti-TGF-ß monoclonal antibody (mAb) treatment on Mreg suppressive capacity were tested from the supernatants of the MLR assays. RESULTS: We demonstrated that induced Mreg with a phenotype of CD14low CD16-/low CD80low CD83-/low CD86+/hi HLA-DR+/hi were capable of suppressing proliferating human PBMC, CD4+, and CD8+ T cells, even at a higher responder:Mreg ratio of 32:1 in a pig-human xenogeneic MLR. The strong suppressive potency of Mreg was further correlated with their upregulated IDO expression and activity. The IDO upregulation of Mreg was associated with an increased production of IFN-γ, an IDO stimulator, by xenoreactive responder cells in the xenogeneic MLR. While no effect on Mreg suppressive potency was detected by addition of the iNOS inhibitor L-NMMA or anti-TGF-ß mAb into the MLR assays, inhibition of IDO activity by neutralizing IFN-γ or by IDO inhibitor 1-MT substantially impaired the capacity of Mreg to suppress the xenogeneic response, indicating the importance of upregulated IDO activity in Mreg-mediated suppression of the xenogeneic response in vitro. CONCLUSION: This study demonstrates that human Mreg are capable of suppressing the xenoimmune response in vitro via IDO-involved mechanism(s), suggesting their potential role as an effective immunomodulatory tool in xenotransplantation.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Terapia de Inmunosupresión , Monocitos/inmunología , Porcinos , Linfocitos T/inmunología , Trasplante Heterólogo/métodos
6.
Xenotransplantation ; 24(6)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28963731

RESUMEN

BACKGROUND: A high immunosuppressive burden is required for long-term islet xenograft survival in non-human primates even using genetically modified donor pigs. AIMS: We aimed to investigate the capacity of baboon regulatory T cells (Treg) to suppress islet xenograft rejection, thereby developing a potential immunoregulatory or tolerance therapy that could be evaluated in NHP models of xenotransplantation. MATERIALS & METHODS: Baboon Treg expanded with stimulation by porcine peripheral blood mononuclear cells (PBMC) were characterized by cell phenotyping and suppressive activity assays in vitro. Their function in vivo was evaluated in neonatal porcine islet cell clusters (NICC) transplanted NOD-SCID IL-2rγ-/- (NSG) mice receiving baboon PBMC alone or with expanded autologous Treg. RESULTS: The majority of expanded Treg coexpressed Foxp3 and CD39 and were highly suppressive of the baboon anti-pig xenogeneic T cell response in vitro. Reconstitution of mice with baboon PBMC alone resulted in NICC xenograft rejection within 35 days. Cotransfer with baboon PBMC and Treg prolonged islet xenograft survival beyond 100 days, correlating with Treg engraftment, intragraft CD39 and Foxp3 gene expression, and reduced graft infiltrating effector T cells and reduced interferon-γ production. DISCUSSION & CONCLUSION: Our data supports the capacity of ex vivo expanded CD39+ baboon Treg to suppress islet xenograft rejection in primatized mice, suggesting it has potential as an adjunctive immunotherapy in preclinical NHP models of xenotransplantation.


Asunto(s)
Xenoinjertos/inmunología , Trasplante de Islotes Pancreáticos , Leucocitos Mononucleares/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD/inmunología , Apirasa/inmunología , Rechazo de Injerto/inmunología , Inmunosupresores/farmacología , Interleucina-2/genética , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Ratones Endogámicos NOD , Ratones SCID , Papio , Porcinos , Trasplante Heterólogo/métodos
7.
Kidney Int ; 90(3): 502-14, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27263492

RESUMEN

Regulatory T cells (Tregs) have been shown to be important in maintaining immune homeostasis and preventing autoimmune disease, including autoimmune kidney disease. It is also likely that they play a role in limiting kidney transplant rejection and potentially in promoting transplant tolerance. Although other subsets of Tregs exist, the most potent and well-defined Tregs are the Foxp3 expressing CD4(+) Tregs derived from the thymus or generated peripherally. These CD4(+)Foxp3(+) Tregs limit autoimmune renal disease in animal models, especially chronic kidney disease, and kidney transplantation. Furthermore, other subsets of Tregs, including CD8 Tregs, may play a role in immunosuppression in kidney disease. The development and protective mechanisms of Tregs in kidney disease and kidney transplantation involve multiple mechanisms of suppression. Here we review the development and function of CD4(+)Foxp3(+) Tregs. We discuss the specific application of Tregs as a therapeutic strategy to prevent kidney disease and to limit kidney transplant rejection and detail clinical trials in this area of transplantation.


Asunto(s)
Rechazo de Injerto/terapia , Enfermedades Renales/cirugía , Trasplante de Riñón/efectos adversos , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Aloinjertos/citología , Aloinjertos/efectos de los fármacos , Aloinjertos/inmunología , Aloinjertos/patología , Animales , Enfermedades Autoinmunes/inmunología , Biopsia , Linfocitos T CD8-positivos/inmunología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Rechazo de Injerto/inmunología , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/uso terapéutico , Riñón/citología , Riñón/inmunología , Riñón/patología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante
8.
Curr Opin Organ Transplant ; 21(6): 595-602, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27805947

RESUMEN

PURPOSE OF REVIEW: Clinical transplant tolerance has been most successfully achieved combining hematopoietic chimerism with kidney transplantation. This review outlines this strategy in animal models and human transplantation, and possible clinical challenges. RECENT FINDINGS: Kidney transplant tolerance has been achieved through chimerism in several centers beginning with Massachusetts General Hospital's success with mixed chimerism in human leukocyte antigen (HLA)-mismatched patients and the Stanford group with HLA-matched patients, and the more recent success of the Northwestern protocol achieving full chimerism. This has challenged the original view that stable mixed chimerism is necessary for organ graft tolerance. However, among the HLA-mismatched kidney transplant-tolerant patients, loss of mixed chimerism does not lead to renal-graft rejection, and the development of host Foxp3+ regulatory T cells has been observed. Recent animal models suggest that graft tolerance through bone marrow chimerism occurs through both clonal deletion and regulatory immune cells. Further, Tregs have been shown to improve chimerism in animal models. SUMMARY: Animal studies continue to suggest ways to improve our current clinical strategies. Advances in chimerism protocols suggest that tolerance may be clinically achievable with relative safety for HLA-mismatched kidney transplants.


Asunto(s)
Células de la Médula Ósea , Trasplante de Riñón , Quimera por Trasplante , Tolerancia al Trasplante , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Antígenos HLA , Humanos
9.
J Hepatol ; 62(5): 1171-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25533663

RESUMEN

BACKGROUND & AIMS: Bile acids (BAs) play a crucial role in dietary fat digestion and in the regulation of lipid, glucose, and energy metabolism. Thyroid-stimulating hormone (TSH) is a hormone produced by the anterior pituitary gland that directly regulates several metabolic pathways. However, the impact of TSH on BA homeostasis remains largely unknown. METHODS: We analyzed serum BA and TSH levels in healthy volunteers under strict control of caloric intake. Thyroidectomized rats were administered thyroxine and injected with different doses of TSH. Tshr(-/-) mice were supplemented with thyroxine, and C57BL/6 mice were injected with Tshr-siRNA via the tail vein. The serum BA levels, BA pool size, and fecal BA excretion rate were measured. The regulation of SREBP-2, HNF-4α, and CYP7A1 by TSH were analyzed using luciferase reporter, RNAi, EMSA, and CHIP assays. RESULTS: A negative correlation was observed between the serum levels of TSH and the serum BA levels in healthy volunteers. TSH administration led to a decrease in BA content and CYP7A1 activity in thyroidectomized rats supplemented with thyroxine. When Tshr was silenced in mice, the BA pool size, fecal BA excretion rate, and serum BA levels all increased. Additionally, we found that HNF-4α acts as a critical molecule through which TSH represses CYP7A1 activity. We further confirmed that the accumulation of mature SREBP-2 protein could impair the capacity of nuclear HNF-4α to bind to the CYP7A1 promoter, a mechanism that appears to mediate the effects of TSH. CONCLUSIONS: TSH represses hepatic BA synthesis via a SREBP-2/HNF-4α/CYP7A1 signaling pathway. This finding strongly supports the notion that TSH is an important pathophysiological regulator of liver BA homeostasis independently of thyroid hormones.


Asunto(s)
Ácidos y Sales Biliares , Colesterol 7-alfa-Hidroxilasa/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Hígado/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Tirotropina/metabolismo , Animales , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/metabolismo , Voluntarios Sanos , Homeostasis/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Teóricos , Ratas , Transducción de Señal/fisiología
11.
Mediators Inflamm ; 2014: 596967, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24803740

RESUMEN

BACKGROUND: Coronary atherosclerotic heart disease (CHD) is one of the major concerns in type 2 diabetes (T2D). The systemic chronic inflammation has been postulated to bridge the increased risk of cardiovascular disease and T2D. We formulated that increased peripheral proinflammatory T helper subsets contributed to the development of cardiovascular complications in diabetic patients. METHODS: The frequencies of peripheral total CD4+ T helper cells, proinflammatory Th1, Th17, and Th22 subsets were determined by flow cytometry in diabetic patients with or without CHD (n = 42 and 67, resp.). RESULTS: Both peripheral frequencies and total numbers of Th1, Th17, and Th22 cells were further increased in diabetic patients with CHD. Logistic regression and categorical cross-table analysis further confirmed that increased proinflammatory Th subsets, especially Th22, were independent risk factors of cardiovascular complication in diabetes. Elevated Th subsets also correlated with increased CRP levels and the atherogenic index of plasma. Moreover, Th1 frequency and Th22 numbers demonstrated remarkable potential in predicting CHD in diabetes. CONCLUSIONS: Increased peripheral proinflammatory T helper subsets act in concert and contribute to the increased prevalence of diabetic cardiovasculopathy. The recently identified Th22 cells might play an independent role in CHD and represent a novel proxy for cardiovascular risks in diabetes.


Asunto(s)
Enfermedad de la Arteria Coronaria/inmunología , Diabetes Mellitus Tipo 2/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Adulto , Anciano , Enfermedad de la Arteria Coronaria/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/metabolismo , Células Th17/metabolismo
12.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 39(8): 818-24, 2014 Aug.
Artículo en Zh | MEDLINE | ID: mdl-25202951

RESUMEN

OBJECTIVE: To establish a method for in vitro expansion of human natural CD4⁺CD25⁺ T regulatory cell (Treg) cells for clinical study and immunotherapy. METHODS: Human natural CD4⁺CD25⁺ Treg were isolated from peripheral blood monocyte cells (PBMCs) by magnetic activated cell sorting (MACS) and expanded by CD3/CD28 expansion beads, IL-2 and rapamycin. The number and the viability of the freshly isolated and expanded Treg were detemined by trypan blue staining. The phenotype and the purity of the freshly isolated and expanded Treg were analyzed by FACS. Treg suppression activity was assessed by mixed lymphocyte reaction (MLR) assay. RESULTS: Human natural Treg were expanded up to 2 000 folds after 3 weeks in culture, and the activity was more than 97%. The expanded Treg retained Treg phenotype as shown by their freshly isolated counterparts, and the purity of CD4⁺CD25⁺FoxP3⁺ Treg was (94.22 ± 2.12)%. The expanded Treg demonstrated a similar potent suppression of both proliferating auto- and allo- CD4⁺CD25⁻ effector T cells in vitro in a cell number-dependent manner. CONCLUSION: An in vitro expansion of human natural Treg was established to obtain large numbers of human Treg with highly suppressive phenotype and function, thereby providing a solution to the availability of sufficient human natural Treg in clinical study and immunotherapy.


Asunto(s)
Técnicas de Cultivo de Célula , Linfocitos T Reguladores/citología , Separación Celular , Células Cultivadas , Humanos , Interleucina-2 , Leucocitos Mononucleares , Prueba de Cultivo Mixto de Linfocitos
13.
JCI Insight ; 9(6)2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38516885

RESUMEN

CD4+Foxp3+ regulatory T cells (Tregs) play an essential role in suppressing transplant rejection, but their role within the graft and heterogeneity in tolerance are poorly understood. Here, we compared phenotypic and transcriptomic characteristics of Treg populations within lymphoid organs and grafts in an islet xenotransplant model of tolerance. We showed Tregs were essential for tolerance induction and maintenance. Tregs demonstrated heterogeneity within the graft and lymphoid organs of tolerant mice. A subpopulation of CD127hi Tregs with memory features were found in lymphoid organs, presented in high proportions within long-surviving islet grafts, and had a transcriptomic and phenotypic profile similar to tissue Tregs. Importantly, these memory-like CD127hi Tregs were better able to prevent rejection by effector T cells, after adoptive transfer into secondary Rag-/- hosts, than naive Tregs or unselected Tregs from tolerant mice. Administration of IL-7 to the CD127hi Treg subset was associated with a strong activation of phosphorylation of STAT5. We proposed that memory-like CD127hi Tregs developed within the draining lymph node and underwent further genetic reprogramming within the graft toward a phenotype that had shared characteristics with other tissue or tumor Tregs. These findings suggested that engineering Tregs with these characteristics either in vivo or for adoptive transfer could enhance transplant tolerance.


Asunto(s)
Linfocitos T Reguladores , Tolerancia al Trasplante , Animales , Ratones , Factores de Transcripción Forkhead , Rechazo de Injerto/prevención & control , Tolerancia Inmunológica , Linfocitos T CD4-Positivos , Subunidad alfa del Receptor de Interleucina-7
14.
Curr Diab Rep ; 13(5): 687-94, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23922060

RESUMEN

Beta cell replacement therapy has been proposed as a novel therapy for the treatment of type 1 diabetes. The proof of concept has been demonstrated with successful islet allotransplantation. Islet xenotransplantation has been proposed as an alternative, more reliable, and infinite source of beta cells. The advantages of islet xenotransplantation are the ability to transplant a well differentiated cell that is responsive to glucose and the potential for genetic modification which focuses the treatment on the donor rather than the recipient. The major hurdle remains overcoming the severe cellular rejection that affects xenografts. This review will focus on the major advances that have occurred with genetic modification and the successful therapeutic strategies that have been demonstrated in nonhuman primates. Novel approaches to overcome cell-mediated rejection including biological agents that target selectively costimulation molecules, the development of local immunosuppression through genetic manipulation, and encapsulation will be discussed. Overall, there has been considerable progress in all these areas, which eventually should lead to clinical trials.


Asunto(s)
Trasplante de Islotes Pancreáticos , Trasplante Heterólogo , Animales , Rechazo de Injerto/inmunología , Xenoinjertos/inmunología , Humanos , Terapia de Inmunosupresión , Inflamación/patología
15.
JCI Insight ; 8(23)2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-37874660

RESUMEN

Regulatory T cells (Tregs) have potential for the treatment of autoimmune diseases and graft rejection. Antigen specificity and functional stability are considered critical for their therapeutic efficacy. In this study, expansion of human Tregs in the presence of porcine PBMCs (xenoantigen-expanded Tregs, Xn-Treg) allowed the selection of a distinct Treg subset, coexpressing the activation/memory surface markers HLA-DR and CD27 with enhanced proportion of FOXP3+Helios+ Tregs. Compared with their unsorted and HLA-DR+CD27+ double-positive (DP) cell-depleted Xn-Treg counterparts, HLA-DR+CD27+ DP-enriched Xn-Tregs expressed upregulated Treg function markers CD95 and ICOS with enhanced suppression of xenogeneic but not polyclonal mixed lymphocyte reaction. They also had less Treg-specific demethylation in the region of FOXP3 and were more resistant to conversion to effector cells under inflammatory conditions. Adoptive transfer of porcine islet recipient NOD/SCID IL2 receptor γ-/- mice with HLA-DR+CD27+ DP-enriched Xn-Tregs in a humanized mouse model inhibited porcine islet graft rejection mediated by 25-fold more human effector cells. The prolonged graft survival was associated with enhanced accumulation of FOXP3+ Tregs and upregulated expression of Treg functional genes, IL10 and cytotoxic T lymphocyte antigen 4, but downregulated expression of effector Th1, Th2, and Th17 cytokine genes, within surviving grafts. Collectively, human HLA-DR+CD27+ DP-enriched Xn-Tregs expressed a specific regulatory signature that enabled identification and isolation of antigen-specific and functionally stable Tregs with potential as a Treg-based therapy.


Asunto(s)
Antígenos HLA-DR , Linfocitos T Reguladores , Ratones , Humanos , Animales , Porcinos , Ratones SCID , Ratones Endogámicos NOD , Antígenos HLA-DR/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo
16.
Hepatobiliary Pancreat Dis Int ; 11(5): 494-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23060394

RESUMEN

BACKGROUND: A major barrier to the clinical application of xenotransplantation as a treatment option for patients is T cell-mediated rejection. Studies based on experimental rodent models of xenograft tolerance or rejection in vivo have provided useful information about the role of T cell immune response in xenotransplantation. However not all observations seen in rodents faithfully recapitulate the human situation. This study aimed to establish a humanized mouse model of xenotransplantation, which mimics xenograft rejection in the context of the human immune system. METHODS: NOD-SCID IL2rgamma-/- mice were transplanted with neonatal porcine islet cell clusters (NICC) followed by reconstitution of human peripheral blood mononuclear cells (PBMC). Human leukocyte engraftment and islet xenograft rejection were confirmed by flow cytometric and histological analyses. RESULTS: In the absence of human PBMC, porcine NICC transplanted into NOD-SCID IL2rgamma-/- mice revealed excellent graft integrity and endocrine function. Human PBMC demonstrated a high level of engraftment in NOD-SCID IL2rgamma-/- mice. Reconstitution of NICC recipient NOD-SCID IL2rgamma-/- mice with human PBMC led to the rapid destruction of NICC xenografts in a PBMC number-dependent manner. CONCLUSIONS: Human PBMC-reconstituted NOD-SCID IL2rgamma-/- mice provide an ideal model to study human immune responses in xenotransplantation. Studies based on this humanized mouse model will provide insight for improving the outcomes of clinical xenotransplantation.


Asunto(s)
Trasplante Heterólogo/inmunología , Animales , Humanos , Subunidad gamma Común de Receptores de Interleucina/fisiología , Trasplante de Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Animales , Porcinos
17.
Front Immunol ; 13: 893985, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35874735

RESUMEN

Porcine islets surviving the acute injury caused by humoral rejection and IBMIR will be subjected to cellular xenograft rejection, which is predominately mediated by CD4+ T cells and is characterised by significant infiltration of macrophages, B cells and T cells (CD4+ and CD8+). Overall, the response is different compared to the alloimmune response and more difficult to suppress. Activation of CD4+ T cells is both by direct and indirect antigen presentation. After activation they recruit macrophages and direct B cell responses. Although they are less important than CD4+ T cells in islet xenograft rejection, macrophages are believed to be a major effector cell in this response. Rodent studies have shown that xenoantigen-primed and CD4+ T cell-activated macrophages were capable of recognition and rejection of pancreatic islet xenografts, and they destroyed a graft via the secretion of various proinflammatory mediators, including TNF-α, reactive oxygen and nitrogen species, and complement factors. B cells are an important mediator of islet xenograft rejection via xenoantigen presentation, priming effector T cells and producing xenospecific antibodies. Depletion and/or inhibition of B cells combined with suppressing T cells has been suggested as a promising strategy for induction of xeno-donor-specific T- and B-cell tolerance in islet xenotransplantation. Thus, strategies that expand the influence of regulatory T cells and inhibit and/or reduce macrophage and B cell responses are required for use in combination with clinical applicable immunosuppressive agents to achieve effective suppression of the T cell-initiated xenograft response.


Asunto(s)
Trasplante de Islotes Pancreáticos , Animales , Antígenos Heterófilos , Rechazo de Injerto , Xenoinjertos , Humanos , Inmunidad Celular , Porcinos , Trasplante Heterólogo
18.
Zhonghua Yi Xue Za Zhi ; 91(30): 2124-8, 2011 Aug 16.
Artículo en Zh | MEDLINE | ID: mdl-22093989

RESUMEN

OBJECTIVE: To employ the technology of interfering RNA (RNAi) to identify the role of Foxp3 in the in vitro suppressive effect of human regulatory T cell (Treg) on effector T cells. METHODS: Expanded human Treg were transfected with siRNA targeting Foxp3 genes. The transfection efficiency, the level of corresponding gene and its protein expression were measured by fluorescent microscopy, fluorescence-activated cell sorting (FACS), real-time PCR and Western blot respectively. The phenotypes of Treg were analyzed by FACS. The siRNA transfected Treg was then co-cultured with porcine PBMC or human PBMC-stimulated autologous CD4+CD25- T cells. Their proliferations were examined by WST-1. Treg and autologous CD4+CD25- T cell-related suppressive cytokines were assessed by ELISA. RESULTS: A 68% transfection efficiency in expanded Treg was achieved for Foxp3 siRNA. Real-time PCR revealed a 61.4% mRNA knockdown induced by siRNA targeting Foxp3 genes in Treg versus the control (P < 0.01). Some Treg-associated surface markers were significantly altered versus the control. And the production of suppressive cytokines was lowered. These changes were correlated with the diminished Treg activity in suppressing the proliferation of effector CD4+CD25- T cells. There was 83% suppression by non-transfected Treg vs 48% suppression by Foxp3 siRNA transfected Treg in xeno-immune response (P < 0.05); and 65% suppression by non-transfected Treg vs 48% suppression by Foxp3 siRNA transfected Treg in allo-immune response (P < 0.01). CONCLUSION: Foxp3 is a key intracellular marker for maintaining the phenotypes and functions of Treg.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , ARN Interferente Pequeño , Linfocitos T Reguladores , Línea Celular , Regulación hacia Abajo , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Humanos
19.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 36(12): 1141-6, 2011 Dec.
Artículo en Zh | MEDLINE | ID: mdl-22246358

RESUMEN

OBJECTIVE: To determine the genetic modification on neonatal porcine islet cell clusters (NICC) by small interfering RNA (siRNA)-mediated tissue factor (TF) knockdown in vitro. METHODS: Porcine NICC were transfected with 5 pairs of designed siRNA respectively or in different combinations with lipofectamine 2000. Transfected NICC were analyzed for TF gene by real-time PCR to select the siRNA which worked best. Meanwhile, the viability of NICC after the TF siRNA transfection was examined by FACS. The efficiency of TF gene and protein suppression was measured by real-time PCR and and FACS respectively. RESULTS: Real-time PCR and FACS showed that a 60% reduction in the TF gene expression and a 50% reduction in the protien level of TF on NICC were achieved by transfecting 3 pairs of selected siRNA. The siRNA transfection had no significant effect on the viability of NICC which was analyzed by FACS. CONCLUSION: The expression of TF on porcine NICC is efficiently suppressed by 3 pairs of designed siRNA in vitro.


Asunto(s)
Técnicas de Silenciamiento del Gen , Islotes Pancreáticos/metabolismo , ARN Interferente Pequeño/genética , Tromboplastina/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Silenciador del Gen , Islotes Pancreáticos/citología , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos , Tromboplastina/metabolismo , Transfección
20.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 36(12): 1134-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22246351

RESUMEN

OBJECTIVE: To ascertain the safety and function of the transplantation of neonatal pig islets (NPIs) for diabetic patients. METHODS: NPIs were injected into the hepatic artery of 22 patients. After the transplantation, the patients were treated with a multiple drug immunosuppressive regimens. The first 14 patients were treated with cyclosporine (CsA), mycophenolate mofetil (MMF) and prednisolon, and porcine C-peptide was not monitored, the following 2 patients were given cyklosporin and MMF only, while the next 6 patients were given a quadruple drug regimen consisting of OKT3, takrolimus, sirolimus and prednisolon. The blood glucose levels,exogenous insulin requirement,HbA1c, porcine endogenous retrovirus (PERV) and liver function were assessed before and after NPI transplantation. The serum porcine C peptide were monitored in last 8 patients. RESULTS: The first 14 patients required less insulin and the HbA1c dropped after the transplantation. In the 2 subsequent patients, the metabolic parameters remained unchanged and monitor of porcine C-peptide was negative. Insulin requirements were reduced in all 6 patients, and HbA1c was normalized 3 months after the transplantation. Significant levels of porcine C-peptide were detected in the patient serum. Two of the patients were given a second injection of NPIs, and one of them became insulin independent for 7 d. No serious adverse events were noted after the transplantation. There was no evidence of PERV transmission. Six out of the 22 patients were followed up for 4-6 years after the NPIs injection, immunosuppressive treatment was stopped 1 year after the transplantation. The patients started to take insulin at the time of follow up. Four patients restricted the intake of sugar, while the other 2 did not. One patient had ketoacidosis twice and slight diabetic retinopathy, and another patient had ketoacidosis induced by acute gastroenteritis. The remaining 4 patients did not have any complications. Assays for PERV were again negative. CONCLUSION: Xenogenic islets can survive and function in the human body. No serious adverse events are noted.


Asunto(s)
Diabetes Mellitus Experimental/cirugía , Diabetes Mellitus Tipo 1/cirugía , Inmunosupresores/uso terapéutico , Trasplante de Islotes Pancreáticos/métodos , Trasplante Heterólogo/métodos , Adolescente , Adulto , Animales , Animales Recién Nacidos , Péptido C/sangre , Perros , Femenino , Estudios de Seguimiento , Arteria Hepática , Humanos , Inyecciones Intraarteriales , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Porcinos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA