Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Transplant ; 24(1): 20-29, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37659605

RESUMO

Strong xenorejection limits the clinical application of porcine islet transplantation in type 1 diabetes. Targeting T cell-mediated rejection is one of the main approaches to improve long-term graft survival. Here we study engraftment and survival of porcine islet cells expressing human programmed cell death ligand-1 (hPD-L1) in a humanized mouse model. Neonatal islet-like clusters (NPICCs) from transgenic hPD-L1 (hPD-L1-Tg) and wild-type (Wt) pigs were transplanted into nonobese diabetic-scid IL2rγnull mice stably reconstituted with human immune cells (hPD-L1 n = 10; Wt n = 6). Primary endpoint was development of normoglycemia during a 16-week observation period after transplantation. Secondary endpoints were porcine C-peptide levels and immune cell infiltration. Animals transplanted with hPD-L1-Tg neonatal islet-like clusters achieved a superior normoglycemic rate (50% versus 0%) and significantly higher plasma C-peptide levels as compared to the Wt group, indicating long-term beta cell function. Intracytoplasmic fluorescence-activated cell sorting analysis and immunohistochemistry revealed significantly decreased frequencies of interferonγ-expressing splenic hCD8-positive T cells and reduced intragraft-infiltrating immune cells. We here demonstrate that expression of hPD-L1 provides strong islet xenograft protection without administration of immunosuppressive drugs. These findings support the hypothesis that hPD-L1 has the capacity to control cellular rejection and therefore represents a very promising transgene candidate for clinical porcine islet xenotransplantation.


Assuntos
Diabetes Mellitus , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Camundongos , Animais , Humanos , Suínos , Antígeno B7-H1/metabolismo , Peptídeo C/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos Knockout , Transplante Heterólogo , Camundongos SCID , Rejeição de Enxerto/etiologia
2.
Xenotransplantation ; 30(2): e12793, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36748727

RESUMO

Neonatal porcine islets (NPIs) are a source of islets for xenotransplantation. In the pig, the pancreatic lobes remain separate, thus, when optimizing NPI isolation, the pancreatic lobes included in the pancreatic digest should be specified. These lobes are the duodenal (DL), splenic (SL) and connecting (CL) lobe that correspond to the head, body-tail, and uncinate process of the human pancreas. In this study we are the first to evaluate all three neonatal porcine pancreatic lobes and NPIs isolated from these lobes. We report, a significant difference in endocrine and progenitor cell composition between lobes, and observed pancreatic duct glands (PDG) within the mesenchyme surrounding exocrine ducts in the DL and CL. Following in vitro differentiation, NPIs isolated from each lobe differed significantly in the percent increase of endocrine cells and final cell composition. Compared to other recipients, diabetic immunodeficient mice transplanted with NPIs isolated from the SL demonstrated euglycemic control as early as 4 weeks (p < 0.05) and achieved normoglycemia by 6 weeks post-transplant (p < 0.01). For the first time we report significant differences between the neonatal porcine pancreatic lobes and demonstrate that NPIs from these lobes differ in xenograft function.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Suínos , Humanos , Camundongos , Transplante Heterólogo , Pâncreas , Células-Tronco
3.
Xenotransplantation ; 29(1): e12720, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34850455

RESUMO

Islet transplantation is a potential treatment option for certain patients with type 1 diabetes; however, it still faces barriers to widespread use, including the lack of tools to monitor islet grafts post-transplantation. This study investigates whether labeling neonatal porcine islets (NPI) with polyvinylpyrrolidone-coated superparamagnetic iron oxide nanoparticles (PVP-SPIO) affects their function, and whether this nanoparticle can be utilized to monitor NPI xenografts with magnetic resonance imaging (MRI) in a mouse model. In vitro, PVP-SPIO-labeled NPI in an agarose gel was visualized clearly by MRI. PVP-SPIO-labeled islets were then transplanted under the kidney capsules of immunodeficient nondiabetic and diabetic mice. All diabetic mice that received transplantation of PVP-SPIO-labeled islets reached normoglycemia. Grafts appeared as hypo-intense areas on MRI and were distinguishable from the surrounding tissues. Following injection of spleen cells from immunocompetent mice, normoglycemic recipient mice became diabetic and islet grafts showed an increase in volume, accompanied by a mixed signal on MRI. Overall, this study demonstrates that PVP-SPIO did not affect the function of NPI that PVP-SPIO-labeled islets were easily seen on MRI, and changes in MRI signals following rejection suggest a potential use of PVP-SPIO-labeled islets to monitor graft viability.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Humanos , Transplante das Ilhotas Pancreáticas/métodos , Nanopartículas Magnéticas de Óxido de Ferro , Imageamento por Ressonância Magnética/métodos , Camundongos , Povidona , Suínos , Transplante Heterólogo/métodos
4.
Xenotransplantation ; 28(3): e12669, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33316848

RESUMO

BACKGROUND: Neonatal porcine islets (NPIs) can restore glucose control in mice, pigs, and non-human primates, representing a potential abundant alternative islet supply for clinical beta cell replacement therapy. However, NPIs are vulnerable to inflammatory insults that could be overcome with genetic modifications. Here, we demonstrate in a series of proof-of-concept experiments the potential of the cytoplasmic ubiquitin-editing protein A20, encoded by the TNFAIP3 gene, as an NPI cytoprotective gene. METHODS: We forced A20 expression in NPI grafts using a recombinant adenovirus 5 (Ad5) vector and looked for impact on TNF-stimulated NF-κB activation and NPI graft function. As adeno-associated vectors (AAV) are clinically preferred vectors but exhibit poor transduction efficacy in NPIs, we next screened a series of AAV serotypes under different transduction protocols for their ability achieve high transduction efficiency and suppress NPI inflammation without impacting NPI maturation. RESULTS: Forcing the expression of A20 in NPI with Ad5 vector blocked NF-κB activation by inhibiting IκBα phosphorylation and degradation, and reduced the induction of pro-inflammatory genes Cxcl10 and Icam1. A20-expressing NPIs also exhibited superior functional capacity when transplanted into diabetic immunodeficient recipient mice, evidenced by a more rapid return to euglycemia and improved GTT compared to unmodified NPI grafts. We found AAV2 combined with a 14-day culture period maximized NPI transduction efficiency (>70% transduction rate), and suppressed NF-κB-dependent gene expression without adverse impact upon NPI maturation. CONCLUSION: We report a new protocol that allows for high-efficiency genetic modification of NPIs, which can be utilized to introduce candidate genes without the need for germline engineering. This approach would be suitable for preclinical and clinical testing of beneficial molecules. We also report for the first time that A20 is cytoprotective for NPI, such that A20 gene therapy could aid the clinical development of NPIs for beta cell replacement.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Animais , Dependovirus , Terapia Genética , Vetores Genéticos , Xenoenxertos , Inflamação , Camundongos , Suínos , Transplante Heterólogo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
5.
Xenotransplantation ; 27(4): e12575, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31814191

RESUMO

BACKGROUND: Neonatal porcine islets (NPIs) are a promising tissue source for clinical islet xenotransplantation. To facilitate graft monitoring and recovery if needed, an extra-hepatic transplant site would be optimal. In addition, islet transplantation into the portal vein has been associated with life-threatening intraperitoneal bleeding, portal vein thrombosis, hepatic steatosis, and loss of islet graft function. Although it is hypoxic, the subcutaneous space is a potential extra-hepatic location for clinical islet transplantation. In this study, we explore the benefits of fibrin scaffolds in enhancing the engraftment and long-term function of NPI grafts in this ectopic site. METHODS: Diabetic immune-compromised mice were transplanted with 5000 NPIs under the kidney capsule (KC), and subcutaneously with or without fibrin (SC + F, SC, respectively). All mice were monitored for reversal of hyperglycemia and long-term metabolic function. RESULTS: All mice transplanted with NPI under the KC or SC + F (12/12 and 17/17, respectively) achieved normal fasting blood glucose levels between 5 and 22 weeks post-transplantation and displayed normal glucose tolerance during an intraperitoneal glucose tolerance test. In contrast, NPIs transplanted SC with no fibrin (n = 7) failed to obtain normoglycemia. CONCLUSION: Fibrin matrix facilitates engraftment of NPIs in the subcutaneous site of diabetic mice. These data support further investigation of the subcutaneous site for clinical islet xenotransplantation.


Assuntos
Diabetes Mellitus Experimental , Fibrina , Transplante das Ilhotas Pancreáticas , Animais , Glicemia , Diabetes Mellitus Experimental/cirurgia , Sobrevivência de Enxerto , Ilhotas Pancreáticas , Camundongos , Suínos , Transplante Heterólogo
6.
Xenotransplantation ; 27(4): e12581, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31930606

RESUMO

BACKGROUND: Cell transplantation has been widely recognized as a curative treatment strategy for variety of diseases including type I diabetes (T1D). Broader patient inclusion for this therapeutic option is restricted by a limited supply of healthy human islet donors and significant loss of islets immediately postintrahepatic transplant due to immune activation. Neonatal porcine islets (NPIs) are a potential ubiquitous ß-cell source for treating T1D. Mesenchymal stem cells (MSCs) have the inherent capacity to secrete immunoregulatory, anti-inflammatory, and proangiogenic factors and, thus, have the potential to improve islet engraftment, survival, and function. METHODS: Herein, we assessed the effect of human adipose-derived MSCs (AdMSCs) on NPI metabolic outcomes in diabetic mice when co-transplanted within the prevascularized subcutaneous deviceless (DL) space or kidney capsule (KC). Graft function has been evaluated by weekly blood glucose, stimulated porcine insulin, glucose tolerance, and total cellular graft insulin content. RESULTS: Compared with NPI alone, co-transplantation of NPIs and AdMSCs resulted in significantly earlier normoglycemia (*P < .05), improved glucose tolerance (*P < .05), superior stimulated serum porcine insulin (**P < .01), and increased graft insulin content (*P < .05) in the DL site and not the KC. CONCLUSIONS: Thus, our study demonstrates that co-transplantation of human AdMSCs with NPIs is an effective tactic to augment islet xenograft function in a clinically relevant extrahepatic site.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Transplante de Células-Tronco Mesenquimais , Animais , Glicemia , Diabetes Mellitus Experimental/cirurgia , Xenoenxertos , Humanos , Insulina , Ilhotas Pancreáticas , Células-Tronco Mesenquimais , Camundongos , Suínos , Transplante Heterólogo
7.
FASEB J ; 32(6): 3242-3253, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29401607

RESUMO

Deficient insulin secretion caused by immaturity is the predominant disadvantage of neonatal porcine islets (NPIs) when they serve as a source for islet xenotransplantation. We hypothesize that the transplantation of NPIs with a combination of mesenchymal stem cells (MSCs) can accelerate NPI maturation and improve the engraftment and function of NPIs. After indirect coculturing with monkey MSCs over 21 d, insulin secretion and the expression of regulatory genes relevant to development were assessed in NPIs. NPIs alone or in combination with allogeneic MSCs were intraportally transplanted into diabetic monkeys. Glycemic control was monitored, and graft function was evaluated. Our results suggest that MSCs benefit both the development and proliferation of NPIs in the coexisting systems in vitro and in vivo. These effects are dependent on platelet-derived growth factor receptor-α and are relevant to the inhibition of downstream target Notch1 signaling and the activation of PI3K/protein kinase B signaling.-He, S., Wang, C., Du, X., Chen, Y., Zhao, J., Tian, B., Lu, H., Zhang, Y., Liu, J., Yang, G., Li, L., Li, H., Cheng, J., Lu, Y. MSCs promote the development and improve the function of neonatal porcine islet grafts.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Aloenxertos , Animais , Animais Recém-Nascidos , Xenoenxertos , Macaca mulatta , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Notch1/metabolismo , Suínos
8.
Xenotransplantation ; 26(6): e12546, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31410915

RESUMO

BACKGROUND: Neonatal pigs have the potential to provide an inexhaustible source of islets for the treatment of type 1 diabetes. However, the immunological barriers to islet xenotransplantation still need to be overcome. A better understanding of the xeno-specific immune responses that are involved in neonatal porcine islet (NPI) xenotransplant rejection will help to facilitate the identification of new targets for anti-rejection therapies, and thus enable more specific targeting of the immune cells and molecules involved. METHODS: In this study, we examined the early events of NPI xenograft rejection in the absence of autoimmunity using an immune-competent B6 mouse transplant model. Immune cells were identified by immunohistochemistry and immune molecules were identified by reverse transcription-PCR and flow cytometry assays. RESULTS: Our results demonstrated that early events in NPI xenograft rejection are characterized by initial infiltration of innate immune cells such as macrophages (M1) and neutrophils. CONCLUSIONS: Targeting these cells, which appear early in the rejection process, may provide an opportunity to abort the rejection process prior to activation of T cells. One strategy could be the blockade of chemotactic signals associated with preferential recruitment of immune cells into the graft site. Collectively, our studies demonstrated that early recruitment of immune cells into graft site is controlled by chemotactic activities and suggest a potential target to prevent the early infiltration of immune cells within the graft. Our findings in this study will have significance in improving NPI xenograft acceptance and induce long-term xenograft survival.


Assuntos
Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Xenoenxertos/imunologia , Transplante das Ilhotas Pancreáticas , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/imunologia , Modelos Animais de Doenças , Transplante das Ilhotas Pancreáticas/métodos , Camundongos , Suínos , Transplante Heterólogo/métodos
9.
Xenotransplantation ; 22(6): 443-50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26490671

RESUMO

BACKGROUND: Intraportal infusion is currently the method of choice for clinical islet cell transplantation but suffers from poor efficacy. As the liver may not represent an optimal transplantation site for Langerhans islets, we examined the potential of neonatal porcine islet-like clusters (NPICCs) to engraft in skeletal muscle as an alternative transplantation site. METHODS: Neonatal porcine islet-like clusters were isolated from 2- to 5-day-old piglets and either transplanted under the kidney capsule (s.k.) or injected into the lower hindlimb muscle (i.m.) of streptozotocin-diabetic NOD-SCID IL2rγ(-/-) (NSG) mice. Survival, vascularization, maturation, and functional activity were analyzed by intraperitoneal glucose tolerance testing and immunohistochemical analyses. RESULTS: Intramuscular transplantation of NPICCs resulted in development of normoglycemia and restored glucose homeostasis. Time to reversal of diabetes and glucose tolerance (AUC glucose and AUC insulin) did not significantly differ as compared to s.k. transplantation. Intramuscular grafts exhibited rapid neovascularization and graft composition with cytokeratin-positive ductal cells and beta cells at post-transplant weeks 2 and 8 and after establishment of normoglycemia was comparable in both groups. CONCLUSIONS: Intramuscular injection represents a minimally invasive but efficient alternative for transplantation of NPICCs and, thus, offers an attractive alternative site for xenotransplantation approaches. These findings may have important implications for improving the outcome and the monitoring of pig islet xenotransplantation.


Assuntos
Diabetes Mellitus Experimental/patologia , Sobrevivência de Enxerto/fisiologia , Insulina/sangue , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Animais , Glicemia/análise , Teste de Tolerância a Glucose/métodos , Transplante das Ilhotas Pancreáticas/métodos , Camundongos Endogâmicos NOD , Camundongos SCID , Suínos , Fatores de Tempo
10.
Front Endocrinol (Lausanne) ; 13: 822191, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35222280

RESUMO

Type 1 diabetes (T1D) is a widespread disease, affecting approximately 41.5 million people worldwide. It is generally treated with exogenous insulin, maintaining physiological blood glucose levels but also leading to long-term therapeutic complications. Pancreatic islet cell transplantation offers a potential alternative treatment to insulin injections. Shortage of human organ donors has raised the interest for porcine islet xenotransplantation. Neonatal porcine islets are highly available, can proliferate and mature in vitro as well as after transplantation in vivo. Despite promising preclinical results, delayed insulin secretion caused by immaturity and immunogenicity of the neonatal porcine islets remains a challenge for their clinical application. Multipotent mesenchymal stromal cells (MSCs) are known to have pro-angiogenic, anti-inflammatory and immunomodulatory effects. The current state of research emphasizes the great potential of co-culture and co-transplantation of islet cells with MSCs. Studies have shown enhanced islet proliferation and maturation, insulin secretion and graft survival, resulting in an improved graft outcome. This review summarizes the immunomodulatory and anti-inflammatory properties of MSC in the context of islet transplantation.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Animais , Sobrevivência Celular , Técnicas de Cocultura , Sobrevivência de Enxerto , Humanos , Imunomodulação , Células-Tronco Mesenquimais/imunologia , Neovascularização Fisiológica , Suínos , Transplante Heterólogo
11.
Polymers (Basel) ; 14(6)2022 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-35335450

RESUMO

The subcutaneous space is currently being pursued as an alternative transplant site for ß-cell replacement therapies due to its retrievability, minimally invasive procedure and potential for graft imaging. However, implantation of ß-cells into an unmodified subcutaneous niche fails to reverse diabetes due to a lack of adequate blood supply. Herein, poly (ε-caprolactone) (PCL) and poly (lactic-co-glycolic acid) (PLGA) polymers were used to make scaffolds and were functionalized with peptides (RGD (Arginine-glycine-aspartate), VEGF (Vascular endothelial growth factor), laminin) or gelatin to augment engraftment. PCL, PCL + RGD + VEGF (PCL + R + V), PCL + RGD + Laminin (PCL + R + L), PLGA and PLGA + Gelatin (PLGA + G) scaffolds were implanted into the subcutaneous space of immunodeficient Rag mice. After four weeks, neonatal porcine islets (NPIs) were transplanted within the lumen of the scaffolds or under the kidney capsule (KC). Graft function was evaluated by blood glucose, serum porcine insulin, glucose tolerance tests, graft cellular insulin content and histologically. PLGA and PLGA + G scaffold recipients achieved significantly superior euglycemia rates (86% and 100%, respectively) compared to PCL scaffold recipients (0% euglycemic) (* p < 0.05, ** p < 0.01, respectively). PLGA scaffolds exhibited superior glucose tolerance (* p < 0.05) and serum porcine insulin secretion (* p < 0.05) compared to PCL scaffolds. Functionalized PLGA + G scaffold recipients exhibited higher total cellular insulin contents compared to PLGA-only recipients (* p < 0.05). This study demonstrates that the bioabsorption of PLGA-based fibrous scaffolds is a key factor that facilitates the function of NPIs transplanted subcutaneously in diabetic mice.

12.
Cell Transplant ; 28(7): 967-972, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31037984

RESUMO

The limited availability of human islets has led to the examination of porcine islets as a source of clinically suitable tissue for transplantation in patients with diabetes mellitus. Islets from porcine donors are commonly used in both in vitro and in vivo experiments studying diabetes mellitus. However, there are significant differences in quality and quantity of islet yield depending on donor pig age, as well as substantial differences in the costs of pancreas procurement in adult versus neonatal and juvenile pigs. In this study, we compared the total cost per islet of juvenile pig pancreata with that of neonatal and adult pigs. Although adult porcine pancreata yield, on average, more than five times the amount of islets than do juvenile and neonatal pancreata, we found that the high price of adult pigs led to the cost per islet being more than twice that of juvenile and neonatal islets (US $0.09 vs $0.04 and $0.02, respectively). In addition, neonatal and juvenile islets are advantageous in their scalability and retention of viability after culture. Our findings indicate that isolating neonatal and juvenile porcine islets is more cost-effective and scalable than isolating adult porcine islets.


Assuntos
Transplante das Ilhotas Pancreáticas/economia , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/cirurgia , Animais , Diabetes Mellitus Tipo 1/cirurgia , Masculino , Suínos , Transplante Heterólogo/economia , Transplante Heterólogo/métodos
13.
J Endocrinol ; 235(3): 237-249, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28931557

RESUMO

Oxidative stress is a major cause of islet injury and dysfunction during isolation and transplantation procedures. Cyanidin-3-O-glucoside (C3G), which is present in various fruits and vegetables especially in Chinese bayberry, shows a potent antioxidant property. In this study, we determined whether C3G could protect neonatal porcine islets (NPI) from reactive oxygen species (H2O2)-induced injury in vitro and promote the function of NPI in diabetic mice. We found that C3G had no deleterious effect on NPI and that C3G protected NPI from damage induced by H2O2 Significantly higher hemeoxygenase-1 (HO1) gene expression was detected in C3G-treated NPI compared to untreated islets before and after transplantation (P < 0.05). Western blot analysis showed a significant increase in the levels of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphatidylinositol 3-kinase (PI3K/Akt) proteins in C3G-treated NPI compared to untreated islets. C3G induced the nuclear translocation of nuclear erythroid 2-related factor 2 (NRF2) and the significant elevation of HO1 protein. Recipients of C3G-treated NPI with or without C3G-supplemented drinking water achieved normoglycemia earlier compared to recipients of untreated islets. Mice that received C3G-treated islets with or without C3G-supplemented water displayed significantly lower blood glucose levels at 5-10 weeks post-transplantation compared to mice that received untreated islets. Mice that received C3G-treated NPI and C3G-supplemented drinking water had significantly (P < 0.05) lower blood glucose levels at 7 and 8 weeks post-transplantation compared to mice that received C3G-treated islets. These findings suggest that C3G has a beneficial effect on NPI through the activation of ERK1/2- and PI3K/AKT-induced NRF2-mediated HO1 signaling pathway.


Assuntos
Animais Recém-Nascidos , Antocianinas/farmacologia , Antioxidantes/farmacologia , Glucosídeos/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Sus scrofa , Animais , Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/análise , Heme Oxigenase-1/genética , Peróxido de Hidrogênio/farmacologia , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/lesões , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Sistema de Sinalização das MAP Quinases , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/análise , Proteína Quinase 3 Ativada por Mitógeno/análise , Fator 2 Relacionado a NF-E2/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/análise , Espécies Reativas de Oxigênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transplante Heterólogo/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA