Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
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.
Nat Commun ; 12(1): 3362, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099692

RESUMO

Diabetes can be caused by an insufficiency in ß-cell mass. Here, we performed a genetic screen in a zebrafish model of ß-cell loss to identify pathways promoting ß-cell regeneration. We found that both folate receptor 1 (folr1) overexpression and treatment with folinic acid, stimulated ß-cell differentiation in zebrafish. Treatment with folinic acid also stimulated ß-cell differentiation in cultures of neonatal pig islets, showing that the effect could be translated to a mammalian system. In both zebrafish and neonatal pig islets, the increased ß-cell differentiation originated from ductal cells. Mechanistically, comparative metabolomic analysis of zebrafish with/without ß-cell ablation and with/without folinic acid treatment indicated ß-cell regeneration could be attributed to changes in the pyrimidine, carnitine, and serine pathways. Overall, our results suggest evolutionarily conserved and previously unknown roles for folic acid and one-carbon metabolism in the generation of ß-cells.


Assuntos
Carbono/metabolismo , Diferenciação Celular/efeitos dos fármacos , Receptor 1 de Folato/metabolismo , Células Secretoras de Insulina/metabolismo , Leucovorina/farmacologia , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Carnitina/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Receptor 1 de Folato/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células Secretoras de Insulina/citologia , Larva/genética , Larva/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Pirimidinas/metabolismo , Suínos , Peixe-Zebra/genética
5.
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
6.
Compr Physiol ; 10(3): 839-878, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32941683

RESUMO

Islet cell replacement therapies represent an effective way to restore physiologic glycemic control in patients with type 1 diabetes (T1DM) and severe hypoglycemia. Despite being able to provide long-term insulin independence, patients still require lifelong immunosuppression, which has myriad detrimental effects including an increased risk for opportunistic infections and some types of cancer. This vital issue precludes widespread application of these therapies as a true cure for T1DM. Encapsulation of islets into immunoisolating/immunoprotective devices provides the potential of abrogating the requisite for lifelong immunosuppression. The field of cellular encapsulation lies at a complex intersection between the areas of chemistry, physics, bioengineering, cell biology, immunology, and clinical medicine. In diabetes, cellular encapsulation has existed for nearly 50 years, nevertheless, a resurgence of interest in the field has been motivated by promising results in small- and large-animal models. Recent studies have demonstrated that long-term diabetes reversal without immunosuppression is indeed routinely achievable. Future researchers interested in exploring cellular encapsulation strategies will require a clear understanding of the basic theoretical and practical principles, guiding this rapidly expanding field. This article will provide essential considerations concerning the physicochemical properties of the most commonly used biomaterials, relevant aspects of the immune response to bioencapsulation, current encapsulation strategies, potential implantation sites for encapsulated cell therapies and, finally, a comprehensive review on the current state of clinical translation. © 2020 American Physiological Society. Compr Physiol 10:839-878, 2020.


Assuntos
Encapsulamento de Células/métodos , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/citologia , Transplante das Ilhotas Pancreáticas/métodos , Animais , Humanos , Células Secretoras de Insulina/transplante
7.
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
8.
JCI Insight ; 4(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31581152

RESUMO

Islet transplantation can restore lost glycemic control in type 1 diabetes subjects but is restricted in its clinical application by a limiting supply of islets and the need for heavy immune suppression to prevent rejection. TNFAIP3, encoding the ubiquitin editing enzyme A20, regulates the activation of immune cells by raising NF-κB signaling thresholds. Here, we show that increasing A20 expression in allogeneic islet grafts resulted in permanent survival for ~45% of recipients, and > 80% survival when combined with subtherapeutic rapamycin. Allograft survival was dependent upon Tregs and was antigen specific, and grafts showed reduced expression of inflammatory factors. Transplantation of islets with A20 containing a loss-of-function variant (I325N) resulted in increased RIPK1 ubiquitination and NF-κB signaling, graft hyperinflammation, and acute allograft rejection. Overexpression of A20 in human islets potently reduced expression of inflammatory mediators, with no impact on glucose-stimulated insulin secretion. Therapeutic administration of A20 raises inflammatory signaling thresholds to favor immune tolerance and promotes islet allogeneic survival. Clinically, this would allow for reduced immunosuppression and support the use of alternate islet sources.


Assuntos
Tolerância Imunológica/fisiologia , Transplante das Ilhotas Pancreáticas , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/fisiologia , Sobrevivência de Enxerto , Humanos , Transplante Homólogo
9.
PLoS One ; 13(11): e0206449, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30419033

RESUMO

Islet transplantation is an established clinical procedure for select patients with type 1 diabetes and severe hypoglycemia to stabilize glycemic control. Post-transplant, substantial beta cell mass is lost, necessitating multiple donors to maintain euglycemia. A potential strategy to augment islet engraftment is the co-transplantation of islets with multipotent mesenchymal stem cells to capitalize upon their pro-angiogenic and anti-inflammatory properties. Herein, we examine the in vitro and in vivo effect of co-culturing murine islets with human adipose-derived mesenchymal stem cells (Ad-MSCs). Islets co-cultured with Ad-MSCs for 48 hours had decreased cell death, superior viability as measured by membrane integrity, improved glucose stimulated insulin secretion and reduced apoptosis compared to control islets. These observations were recapitulated with human islets, albeit tested in a limited capacity. Recipients of marginal mouse islet mass grafts, co-transplanted with Ad-MSCs without a co-culture period, did not reverse to normoglycemia as efficiently as islets alone. However, utilizing a 48-hour co-culture period, marginal mouse islets grafts with Ad-MSCs achieved a superior percent euglycemia rate when compared to islets cultured and transplanted alone. A co-culture period of human islets with human Ad-MSCs may have a clinical benefit improving engraftment outcomes.


Assuntos
Tecido Adiposo/citologia , Técnicas de Cocultura , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Feminino , Humanos , Rim/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Tempo
10.
Diabetes ; 66(5): 1312-1321, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28246290

RESUMO

Mesenchymal stem cells (MSCs) possess immunoregulatory, anti-inflammatory, and proangiogenic properties and, therefore, have the potential to improve islet engraftment and survival. We assessed the effect human bone marrow-derived MSCs have on neonatal porcine islets (NPIs) in vitro and determined islet engraftment and metabolic outcomes when cotransplanted in a mouse model. NPIs cocultured with MSCs had greater cellular insulin content and increased glucose-stimulated insulin secretion. NPIs were cotransplanted with or without MSCs in diabetic B6.129S7-Rag1tm1Mom/J mice. Blood glucose and weight were monitored until reversal of diabetes; mice were then given an oral glucose tolerance test. Islet grafts were assessed for the degree of vascularization and total cellular insulin content. Cotransplantation of NPIs and MSCs resulted in significantly earlier normoglycemia and vascularization, improved glucose tolerance, and increased insulin content. One experiment conducted with MSCs from a donor with an autoimmune disorder had no positive effects on transplant outcomes. Cotransplantation of human MSCs with NPIs demonstrated a beneficial metabolic effect likely as a result of earlier islet vascularization and improved islet engraftment. In addition, donor pathology of MSCs can influence the functional capacity of MSCs.


Assuntos
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Transplante de Células-Tronco Mesenquimais , Neovascularização Fisiológica , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/irrigação sanguínea , Masculino , Camundongos , Suínos , Transplantes/irrigação sanguínea
11.
Cell Transplant ; 25(2): 327-42, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26109574

RESUMO

We previously demonstrated that short-term administration of a combination of anti-LFA-1 and anti-CD154 monoclonal antibodies (mAbs) induces tolerance to neonatal porcine islet (NPI) xenografts that is mediated by regulatory T cells (Tregs) in B6 mice. In this study, we examined whether the coinhibitory molecule PD-1 is required for the induction and maintenance of tolerance to NPI xenografts. We also determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or xenogeneic rat islet grafts since we previously demonstrated that tolerance to NPI xenografts could be extended to second-party NPI xenografts. Finally, we determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or second-party porcine skin grafts. Diabetic B6 mice were transplanted with 2,000 NPIs under the kidney capsule and treated with short-term administration of a combination of anti-LFA-1 and anti-CD154 mAbs. Some of these mice were also treated simultaneously with anti-PD-1 mAb at >150 days posttransplantation. Spleen cells from some of the tolerant B6 mice were used for proliferation assays or were injected into B6 rag(-/-) mice with established islet grafts from allogeneic or xenogeneic donors. All B6 mice treated with anti-LFA-1 and anti-CD154 mAbs achieved and maintained normoglycemia until the end of the study; however, some mice that were treated with anti-PD-1 mAb became diabetic. All B6 rag(-/-) mouse recipients of first- and second-party NPIs maintained normoglycemia after reconstitution with spleen cells from tolerant B6 mice, while all B6 rag(-/-) mouse recipients of allogeneic mouse or xenogeneic rat islets rejected their grafts after cell reconstitution. Tolerant B6 mice rejected their allogeneic mouse or xenogeneic second-party porcine skin grafts while remaining normoglycemic until the end of the study. These results show that porcine islet-specific tolerance is dependent on PD-1, which could not be extended to skin grafts.


Assuntos
Anticorpos Monoclonais/farmacologia , Diabetes Mellitus/imunologia , Sobrevivência de Enxerto/fisiologia , Tolerância Imunológica/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Transplante de Pele , Animais , Ligante de CD40/imunologia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Antígeno-1 Associado à Função Linfocitária/imunologia , Masculino , Camundongos Knockout , Transplante de Pele/métodos , Suínos , Transplante Heterólogo/métodos
12.
Cardiovasc Res ; 101(2): 306-16, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24259499

RESUMO

AIMS: Methylglyoxal (MG) accumulates in diabetes and impairs neovascularization. This study assessed whether overexpressing the MG-metabolizing enzyme glyoxalase-1 (GLO1) in only bone marrow cells (BMCs) could restore neovascularization in ischaemic tissue of streptozotocin-induced diabetic mice. METHODS AND RESULTS: After 24 h of hyperglycaemic and hypoxic culture, BMCs from GLO1 overexpressing and wild-type (WT) diabetic mice were compared for migratory potential, viability, and mRNA expression of anti-apoptotic genes (Bcl-2 and Bcl-XL). In vivo, BMCs from enhanced green fluorescent protein (eGFP) mice that overexpress GLO1 were used to reconstitute the BM of diabetic mice (GLO1-diabetics). Diabetic and non-diabetic recipients of WT GFP(+) BM served as controls (WT-diabetics and non-diabetics, respectively). Following hindlimb ischaemia, the mobilization of BMCs was measured by flow cytometry. In hindlimbs, the presence of BM-derived angiogenic (GFP(+)CXCR4(+)) and endothelial (GFP(+)vWF(+)) cells and also arteriole density were determined by immunohistochemistry. Hindlimb perfusion was measured using laser Doppler. GLO1-BMCs had superior migratory potential, increased viability, and greater Bcl-2 and Bcl-XL expression, compared with WT BMCs. In vivo, the mobilization of pro-angiogenic BMCs (CXCR4(+), c-kit(+), and Flk(+)) was enhanced post-ischaemia in GLO1-diabetics compared to WT-diabetics. A greater number of GFP(+)CXCR4(+) and GFP(+)vWF(+) BMCs incorporated into the hindlimb tissue of GLO1-diabetics and non-diabetics than in WT-diabetics. Arteriole and capillary density and perfusion were also greater in GLO1-diabetics and non-diabetics. CONCLUSION: This study demonstrates that protection from MG uniquely in BM is sufficient to restore BMC function and neovascularization of ischaemic tissue in diabetes and identifies GLO1 as a potential therapeutic target.


Assuntos
Células da Medula Óssea/enzimologia , Transplante de Medula Óssea , Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 1/cirurgia , Angiopatias Diabéticas/cirurgia , Isquemia/cirurgia , Lactoilglutationa Liase/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Patológica , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Apoptose , Glicemia/metabolismo , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatologia , Angiopatias Diabéticas/enzimologia , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/fisiopatologia , Membro Posterior , Humanos , Isquemia/enzimologia , Isquemia/genética , Isquemia/fisiopatologia , Lactoilglutationa Liase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/patologia , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Oxirredução , Estresse Oxidativo , Aldeído Pirúvico/metabolismo , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Fatores de Tempo , Regulação para Cima
13.
Biol Reprod ; 90(1): 13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24285718

RESUMO

Conventionally, it was believed that Sertoli cells (SC) stopped proliferating at puberty and became terminally differentiated quiescent cells. However, recent studies have challenged that dogma. In this study, we transplanted nondividing SC isolated from 23- to 27-day-old postpubertal rats transduced with a recombinant adenoviral vector (containing furin-modified human proinsulin cDNA) into diabetic severe combined immunodeficiency mice. Immunostaining the grafts for cell proliferation markers, proliferating cell nuclear antigen (PCNA) and MKI67, revealed that transplanted SC within the grafts were proliferating. Possible causes for resumption of proliferation of SC could be viral transduction, cell isolation and culture, higher abdominal temperature at the transplant site, and/or transplantation. To test for these possible causes, double- immunofluorescence staining was performed for GATA4 (SC marker) and MKI67. None of the SC were positive for MKI67 in tissue collected during SC isolation and culture or at higher temperature. However, nontransduced SC stained positive for MKI67 after transplantation into rats, suggesting viral transduction was not a key factor for induction of SC proliferation. Interestingly, resumption in proliferative ability of nondividing SC was temporary, as SC stopped proliferating within 14 days of transplantation and did not proliferate thereafter. Quantification of 5-bromo-2'-deoxyuridine-labeled SC demonstrated that 7%-9% of the total transplanted SC were proliferating in the grafts. These data indicate for the first time that nondividing SC resumed proliferation after transplantation and further validate previous findings that SC are not terminally differentiated. Hence, transplantation of SC could provide a useful model with which to study the regulation of SC proliferation in vivo.


Assuntos
Proliferação de Células , Células de Sertoli/citologia , Células de Sertoli/fisiologia , Células de Sertoli/transplante , Maturidade Sexual/fisiologia , Animais , Divisão Celular , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ratos , Ratos Endogâmicos Lew , Ratos Endogâmicos WF
14.
PLoS One ; 7(5): e38189, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22666480

RESUMO

Transplantation of human islets is an attractive alternative to daily insulin injections for patients with type 1 diabetes. However, the majority of islet recipients lose graft function within five years. Inflammation is a primary contributor to graft loss, and inhibiting pro-inflammatory cytokine activity can reverse inflammation mediated dysfunction of islet grafts. As mesenchymal stem cells (MSCs) possess numerous immunoregulatory properties, we hypothesized that MSCs could protect human islets from pro-inflammatory cytokines. Five hundred human islets were co-cultured with 0.5 or 1.0 × 10(6) human MSCs derived from bone marrow or pancreas for 24 hours followed by 48 hour exposure to interferon-γ, tumor necrosis factor-α and interleukin 1ß. Controls include islets cultured alone (± cytokines) and with human dermal fibroblasts (± cytokines). For all conditions, glucose stimulated insulin secretion (GSIS), total islet cellular insulin content, islet ß cell apoptosis, and potential cytoprotective factors secreted in the culture media were determined. Cytokine exposure disrupted human islet GSIS based on stimulation index and percentage insulin secretion. Conversely, culture with 1.0 × 10(6) bMSCs preserved GSIS from cytokine treated islets. Protective effects were not observed with fibroblasts, indicating that preservation of human islet GSIS after exposure to pro-inflammatory cytokines is MSC dependent. Islet ß cell apoptosis was observed in the presence of cytokines; however, culture of bMSCs with islets prevented ß cell apoptosis after cytokine treatment. Hepatocyte growth factor (HGF) as well as matrix metalloproteinases 2 and 9 were also identified as putative secreted cytoprotective factors; however, other secreted factors likely play a role in protection. This study, therefore, demonstrates that MSCs may be beneficial for islet engraftment by promoting cell survival and reduced inflammation.


Assuntos
Citocinas/farmacologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Adolescente , Adulto , Idoso , Apoptose/efeitos dos fármacos , Células da Medula Óssea/citologia , Agregação Celular , Técnicas de Cocultura , Citoproteção/efeitos dos fármacos , Glucose/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Inflamação/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Pessoa de Meia-Idade , Adulto Jovem
15.
Islets ; 4(3): 207-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22688061

RESUMO

We report on the incidental observation of ectopic pancreas in a donor for islet cell transplantation. The donor's clinical and imaging presentation was definitive for holoprosencephaly. This case report discusses a possible link between ectopic pancreas and holoprosencephaly.


Assuntos
Coristoma/complicações , Duodenopatias/complicações , Holoprosencefalia/complicações , Pâncreas , Coristoma/patologia , Duodenopatias/patologia , Evolução Fatal , Holoprosencefalia/patologia , Humanos , Imuno-Histoquímica , Masculino , Adulto Jovem
17.
Diabetes ; 59(4): 958-66, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20086231

RESUMO

OBJECTIVE: The objective of this study was to determine whether tolerance to neonatal porcine islet (NPI) xenografts could be achieved by short-term administrations of anti-LFA-1 and anti-CD154 monoclonal antibodies (mAbs). RESEARCH DESIGN AND METHODS: Diabetic B6 mice received NPI transplants and short-term injections of combined anti-LFA-1 and anti-CD154 mAbs. Mice with long-term islet graft function were treated with depleting anti-CD25 mAb or re-transplanted with a second-party NPI. At the end of the study, grafts from mice with long-term islet function were examined. Their spleen cells were characterized and used for in vitro proliferation and adoptive transfer studies. RESULTS: All mAb-treated NPI recipients maintained normoglycemia for >100 days post-transplantation. Only 5 of 50 mice rejected their grafts before 300 days post-transplantation. Intact islets, foxp3(+) immune cells, as well as interleukin (IL)-10 and transforming growth factor (TGF)-beta regulatory cytokine transcripts were detected in the NPI xenografts from tolerant mice. A higher percentage of CD4(+) T-cell population from these mice expressed regulatory markers, suggesting that tolerance to NPI xenografts may be mediated by T regulatory cells. This was confirmed when tolerant mice treated with depleting anti-CD25 mAb became diabetic. Lymphocytes from tolerant mice inhibited the proliferation of lymphocytes from B6 mice immunized with porcine cells and they displayed limited proliferation when adoptively transferred. All protected B6 mice transplanted with a second-party NPI xenograft maintained long-term normoglycemia even after removal of the first NPI graft-bearing kidney. CONCLUSIONS: These results demonstrate that tolerance to NPI xenografts can be achieved by transient administrations of combined anti-LFA-1 and anti-CD154 mAb therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Ligante de CD40/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Tolerância ao Transplante/imunologia , Transplante Heterólogo/imunologia , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Imunofenotipagem , Camundongos , Nefrectomia , Suínos
18.
Neonatology ; 96(3): 162-70, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19332996

RESUMO

BACKGROUND: Neonatal asphyxia may lead to the development of ischemia-reperfusion induced intestinal injury, which is related to oxygen-derived free radical production. N-Acetylcysteine (NAC) is a thiol-containing antioxidant which increases intracellular stores of glutathione. OBJECTIVES: Using a swine model of neonatal hypoxia-reoxygenation, we examined whether administration of NAC after resuscitation improved intestinal perfusion and reduced intestinal damage. METHODS: Twenty-four piglets (1-4 days old, 1.4-2.2 kg) were anesthetized and acutely instrumented for continuous monitoring of superior mesenteric arterial flow and oxygen delivery. Alveolar hypoxia was induced for 2 h, followed by resuscitation with 100% oxygen for 1 h and 21% oxygen for 3 h. Animals were randomized to sham-operated, hypoxic control and NAC treatment (150 mg/kg i.v. at 0 or 10 min of reoxygenation followed by infusion 100 mg/kg/h) groups. During hypoxia-reoxygenation, intestinal tissue glutathione content, caspase-3 activity and reoxygenation injury were examined. RESULTS: After 2 h of hypoxia, piglets were acidotic and hypotensive, with significantly depressed blood flow and oxygen delivery to the small intestine. Upon reoxygenation, hemodynamics recovered as did oxygen supply to the small intestine. After 4 h of reoxygenation, the NAC treatment improved mesenteric flow and oxygen delivery. Despite reducing the increase in caspase-3 activities after hypoxia-reoxygenation by NAC treatment, no significant differences in the glutathione content and histological grading of ileal injury were found among the experimental groups. CONCLUSIONS: In newborn piglets with hypoxia-reoxygenation, NAC may improve mesenteric blood flow and oxygen delivery without significant effect on tissue glutathione content. The protective role of NAC in the reoxygenated intestine after severe hypoxia warrants further investigation.


Assuntos
Acetilcisteína/uso terapêutico , Antioxidantes/uso terapêutico , Enterocolite/tratamento farmacológico , Hipóxia/tratamento farmacológico , Oxigênio/efeitos adversos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Enterocolite/metabolismo , Enterocolite/patologia , Hipóxia/complicações , Hipóxia/metabolismo , Íleo/efeitos dos fármacos , Íleo/patologia , Oxigenoterapia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Circulação Esplâncnica/efeitos dos fármacos , Suínos
19.
Shock ; 31(3): 292-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18650774

RESUMO

Neonatal asphyxia can result in poor perfusion, vasoconstriction, and decreased oxygen delivery in the intestine. Milrinone increases myocardial contractility and causes peripheral vasodilatation. We examined the dose-response of milrinone on the intestinal circulation, oxygen metabolism, and injury in a newborn piglet model of asphyxia-reoxygenation. Piglets (aged 1-3 days, weighing 1.5-2.3 kg) were acutely instrumented to measure superior mesenteric artery (SMA) flow and oxygen delivery. After stabilization, hypoxia (inspired oxygen concentration, 0.08-0.15) was induced for 2 h followed by reoxygenation with 100% O2 for 1 h then 21% O2 for 3 h. At 2 h of reoxygenation, saline or milrinone infusion at doses of 0.25, 0.5, or 0.75 microg/kg per min was given for 2 h in a blinded randomized fashion (n = 7 per group). Hemodynamic and oxygen transport parameters were analyzed at predefined time points. Intestinal tissue lactate concentrations, plasma milrinone levels, and intestinal glutathione redox status were determined at the end of the experiment. In the intestinal tract, milrinone significantly increased SMA flow and oxygen delivery while decreasing vascular resistance at a dose of 0.75 microg/kg per min (P < 0.05, ANOVA). A modest increase in SMA flow and oxygen delivery was found with milrinone at 0.5 microg/kg per min. Plasma milrinone levels correlated with SMA flow and vascular resistance (r = 0.5 and r = -0.6, respectively, P < 0.05). Intestinal lactate concentrations and histopathology were not significantly different among groups. Oxidized glutathione correlated with SMA vascular resistance and negatively with milrinone levels (r = 0.6 and r = -0.5, P < 0.05). When used to treat shock in a newborn model of asphyxia-reoxygenation, milrinone dose-dependently increases SMA flow and oxygen delivery with a significantly decreased SMA vascular resistance at higher doses.


Assuntos
Asfixia Neonatal/tratamento farmacológico , Cardiotônicos/farmacologia , Intestinos/irrigação sanguínea , Milrinona/farmacologia , Oxigênio/metabolismo , Vasodilatação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Asfixia Neonatal/metabolismo , Asfixia Neonatal/patologia , Asfixia Neonatal/fisiopatologia , Cardiotônicos/farmacocinética , Relação Dose-Resposta a Droga , Glutationa/metabolismo , Humanos , Recém-Nascido , Mucosa Intestinal/metabolismo , Intestinos/fisiopatologia , Ácido Láctico/metabolismo , Artéria Mesentérica Superior/metabolismo , Artéria Mesentérica Superior/patologia , Artéria Mesentérica Superior/fisiopatologia , Milrinona/farmacocinética , Contração Miocárdica/efeitos dos fármacos , Suínos , Fatores de Tempo , Resistência Vascular/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
20.
Lab Invest ; 89(2): 110-21, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19079324

RESUMO

Pancreatic mesenchymal stem cells (MSCs) may be derived from human beta-cells undergoing reversible epithelial-mesenchymal transition (EMT), suggesting that they could be a potential source of new beta-cells. In this study we sought to determine the origin of pancreatic MSCs in the nonendocrine pancreas. Double immunofluorescent (IF) staining and flow cytometry were used to assess the cell phenotype of nonendocrine pancreas tissue following islet procurement, during in vitro expansion of MSCs, and after differentiation. IF staining of paraffin-embedded pancreatic biopsy sections was used to assess cell phenotype in vivo. In this study we demonstrated that: (1) pancreatic epithelial cells do not express MSC antigens in vivo; (2) following islet isolation EpCAM- and CK19-positive epithelial cells coexpressed the MSC antigens CD44 (32+/-8% and 38+/-10%) and CD29 (85+/-4% and 64+/-4%); (3) during in vitro expansion the number of single-positive epithelial and double-positive epithelial/MSCs decreased whereas the number of single-positive MSCs increased and (4) differentiated MSCs do not revert to a true epithelial cell phenotype in our culture conditions, as epithelial cell surface markers (EpCAM, CK19 and E-Cadherin) are not reexpressed, although the MSC phenotype is altered. This study demonstrates that MSCs may be derived in vitro via a pancreatic epithelial cell undergoing EMT, however it is more likely that a small percentage of MSCs that reside in the adult pancreas are proliferating whereas the epithelial cells are negatively selected by the experimental culture conditions.


Assuntos
Artefatos , Células Epiteliais/citologia , Mesoderma/citologia , Pâncreas Exócrino/citologia , Adulto , Idoso , Biomarcadores/metabolismo , Contagem de Células , Técnicas de Cultura de Células , Diferenciação Celular , Células Epiteliais/metabolismo , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Receptores de Hialuronatos/metabolismo , Imunofenotipagem , Integrina beta1/metabolismo , Mesoderma/metabolismo , Microscopia de Fluorescência , Pessoa de Meia-Idade , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA