Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Xenotransplantation ; 30(2): e12793, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36748727

RESUMEN

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.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Porcinos , Humanos , Ratones , Trasplante Heterólogo , Páncreas , Células Madre
2.
Xenotransplantation ; 28(3): e12669, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33316848

RESUMEN

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.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Dependovirus , Terapia Genética , Vectores Genéticos , Xenoinjertos , Inflamación , Ratones , Porcinos , Trasplante Heterólogo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
3.
Xenotransplantation ; 28(6): e12706, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34245064

RESUMEN

BACKGROUND: Islet transplantation with neonatal porcine islets (NPIs) is a promising treatment for type 1 diabetes (T1D), but immune rejection poses a major hurdle for clinical use. Innate immune-derived reactive oxygen species (ROS) synthesis can facilitate islet xenograft destruction and enhance adaptive immune responses. METHODS: To suppress ROS-mediated xenograft destruction, we utilized nanothin encapsulation materials composed of multilayers of tannic acid (TA), an antioxidant, and a neutral polymer, poly(N-vinylpyrrolidone) (PVPON). We hypothesized that (PVPON/TA)-encapsulated NPIs will maintain euglycemia and dampen proinflammatory innate immune responses following xenotransplantation. RESULTS: (PVPON/TA)-encapsulated NPIs were viable and glucose-responsive similar to non-encapsulated NPIs. Transplantation of (PVPON/TA)-encapsulated NPIs into hyperglycemic C57BL/6.Rag or NOD.Rag mice restored euglycemia, exhibited glucose tolerance, and maintained islet-specific transcription factor levels similar to non-encapsulated NPIs. Gene expression analysis of (PVPON/TA)-encapsulated grafts post-transplantation displayed reduced proinflammatory Ccl5, Cxcl10, Tnf, and Stat1 while enhancing alternatively activated macrophage Retnla, Arg1, and Stat6 mRNA accumulation compared with controls. Flow cytometry analysis demonstrated significantly reduced innate immune infiltration, MHC-II, co-stimulatory molecule, and TNF expression with concomitant increases in arginase-1+ macrophages and dendritic cells. Similar alterations in immune responses were observed following xenotransplantation into immunocompetent NOD mice. CONCLUSION: Our data suggest that (PVPON/TA) encapsulation of NPIs is an effective strategy to decrease inflammatory innate immune signals involved in NPI xenograft responses through STAT1/6 modulation without compromising islet function.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Porcinos , Taninos , Trasplante Heterólogo
4.
Am J Transplant ; 20(3): 714-725, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31650674

RESUMEN

The broad application of ß cell transplantation for type 1 diabetes is hindered by the requisite of lifelong systemic immunosuppression. This study examines the utility of localized islet graft drug delivery to subvert the inflammatory and adaptive immune responses. Herein, we have developed and characterized dexamethasone (Dex) eluting Food and Drug Administration-approved micro-Poly(lactic-co-glycolic acid) micelles and examined their efficacy in a fully major histocompatibility complex-mismatch murine islet allograft model. A clinically relevant dose of 46.6 ± 2.8 µg Dex per graft was confirmed when 2 mg of micelles was implemented. Dex-micelles + CTLA-4-Ig (n = 10) resulted in prolonged allograft function with 80% of the recipients demonstrating insulin independence for 60 days posttransplant compared to 40% in empty micelles + CTLA-4-Ig recipients (n = 10, P = .06). Recipients of this combination therapy (n = 8) demonstrated superior glucose tolerance profiles, compared to empty micelles + CTLA-4-Ig recipients (n = 4, P < .05), and significantly reduced localized intragraft proinflammatory cytokine expression. Histologically, increased insulin positive and FOXP3+ T cells were observed in Dex-micelles + CTLA-4-Ig grafts compared to empty micelles + CTLA-4-Ig grafts (P < .01 and P < .05, respectively). Localized drug delivery via micelles elution has the potential to alter the inflammatory environment, enhances allograft survival, and may be an important adjuvant approach to improve clinical islet transplantation outcomes.


Asunto(s)
Trasplante de Islotes Pancreáticos , Micelas , Aloinjertos , Animales , Dexametasona , Supervivencia de Injerto , Ratones , Ratones Endogámicos BALB C
5.
Xenotransplantation ; 27(4): e12575, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31814191

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental , Fibrina , Trasplante de Islotes Pancreáticos , Animales , Glucemia , Diabetes Mellitus Experimental/cirugía , Supervivencia de Injerto , Islotes Pancreáticos , Ratones , Porcinos , Trasplante Heterólogo
6.
Xenotransplantation ; 27(4): e12581, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31930606

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental , Trasplante de Islotes Pancreáticos , Trasplante de Células Madre Mesenquimatosas , Animales , Glucemia , Diabetes Mellitus Experimental/cirugía , Xenoinjertos , Humanos , Insulina , Islotes Pancreáticos , Células Madre Mesenquimatosas , Ratones , Porcinos , Trasplante Heterólogo
7.
Xenotransplantation ; 25(6): e12432, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30052287

RESUMEN

BACKGROUND: There is currently a shortage of human donor pancreata which limits the broad application of islet transplantation as a treatment for type 1 diabetes. Porcine islets have demonstrated potential as an alternative source, but a study evaluating islets from different donor ages under unified protocols has yet to be conducted. METHODS: Neonatal porcine islets (NPI; 1-3 days), juvenile porcine islets (JPI; 18-21 days), and adult porcine islets (API; 2+ years) were compared in vitro, including assessments of oxygen consumption rate, membrane integrity determined by FDA/PI staining, ß-cell proliferation, dynamic glucose-stimulated insulin secretion, and RNA sequencing. RESULTS: Oxygen consumption rate normalized to DNA was not significantly different between ages. Membrane integrity was age dependent, and API had the highest percentage of intact cells. API also had the highest glucose-stimulated insulin secretion response during a dynamic insulin secretion assay and had 50-fold higher total insulin content compared to NPI and JPI. NPI and JPI had similar glucose responsiveness, ß-cell percentage, and ß-cell proliferation rate. Transcriptome analysis was consistent with physiological assessments. API transcriptomes were enriched for cellular metabolic and insulin secretory pathways, while NPI exhibited higher expression of genes associated with proliferation. CONCLUSIONS: The oxygen demand, membrane integrity, ß-cell function and proliferation, and transcriptomes of islets from API, JPI, and NPI provide a comprehensive physiological comparison for future studies. These assessments will inform the optimal application of each age of porcine islet to expand the availability of islet transplantation.


Asunto(s)
Supervivencia de Injerto/inmunología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Consumo de Oxígeno/fisiología , Animales , Animales Recién Nacidos , Diabetes Mellitus Experimental/terapia , Rechazo de Injerto/inmunología , Células Secretoras de Insulina/inmunología , Trasplante de Islotes Pancreáticos/métodos , Páncreas/inmunología , Páncreas/metabolismo , Porcinos , Transcriptoma/inmunología , Trasplante Heterólogo/métodos
8.
Pharmaceutics ; 15(4)2023 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-37111623

RESUMEN

Beta cell replacement therapies can restore glycemic control to select individuals living with type 1 diabetes. However, the obligation of lifelong immunosuppression restricts cell therapies from replacing exogenous insulin administration. Encapsulation strategies can reduce the inherent adaptive immune response; however, few are successfully translated into clinical testing. Herein, we evaluated if the conformal coating of islets with poly(N-vinylpyrrolidone) (PVPON) and tannic acid (TA) (PVPON/TA) could preserve murine and human islet function while conferring islet allograft protection. In vitro function was evaluated using static glucose-stimulated insulin secretion, oxygen consumption rates, and islet membrane integrity. In vivo function was evaluated by transplanting human islets into diabetic immunodeficient B6.129S7-Rag1tm1Mom/J (Rag-/-) mice. The immunoprotective capacity of the PVPON/TA-coating was assessed by transplanting BALB/c islets into diabetic C57BL/6 mice. Graft function was evaluated by non-fasting blood glucose measurements and glucose tolerance testing. Both coated and non-coated murine and human islets exhibited indistinguishable in vitro potency. PVPON/TA-coated and control human islets were able to restore euglycemia post-transplant. The PVPON/TA-coating as monotherapy and adjuvant to systemic immunosuppression reduced intragraft inflammation and delayed murine allograft rejection. This study demonstrates that PVPON/TA-coated islets may be clinically relevant as they retain their in vitro and in vivo function while modulating post-transplant immune responses.

9.
Pharmaceutics ; 15(9)2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37765170

RESUMEN

One strategy to prevent islet rejection is to create a favorable immune-protective local environment at the transplant site. Herein, we utilize localized cyclosporine A (CsA) delivery to islet grafts via poly(lactic-co-glycolic acid) (PLGA) microparticles to attenuate allograft rejection. CsA-eluting PLGA microparticles were prepared using a single emulsion (oil-in-water) solvent evaporation technique. CsA microparticles alone significantly delayed islet allograft rejection compared to islets alone (p < 0.05). Over 50% (6/11) of recipients receiving CsA microparticles and short-term cytotoxic T lymphocyte-associated antigen 4-Ig (CTLA4-Ig) therapy displayed prolonged allograft survival for 214 days, compared to 25% (2/8) receiving CTLA4-Ig alone. CsA microparticles alone and CsA microparticles + CTLA4-Ig islet allografts exhibited reduced T-cell (CD4+ and CD8+ cells, p < 0.001) and macrophage (CD68+ cells, p < 0.001) infiltration compared to islets alone. We observed the reduced mRNA expression of proinflammatory cytokines (IL-6, IL-10, INF-γ, and TNF-α; p < 0.05) and chemokines (CCL2, CCL5, CCL22, and CXCL10; p < 0.05) in CsA microparticles + CTLA4-Ig allografts compared to islets alone. Long-term islet allografts contained insulin+ and intra-graft FoxP3+ T regulatory cells. The rapid rejection of third-party skin grafts (C3H) in islet allograft recipients suggests that CsA microparticles + CTLA4-Ig therapy induced operational tolerance. This study demonstrates that localized CsA drug delivery plus short-course systemic immunosuppression promotes an immune protective transplant niche for allogeneic islets.

10.
Polymers (Basel) ; 14(6)2022 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-35335450

RESUMEN

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.

11.
Lab Invest ; 89(2): 110-21, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19079324

RESUMEN

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.


Asunto(s)
Artefactos , Células Epiteliales/citología , Mesodermo/citología , Páncreas Exocrino/citología , Adulto , Anciano , Biomarcadores/metabolismo , Recuento de Células , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Epiteliales/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Receptores de Hialuranos/metabolismo , Inmunofenotipificación , Integrina beta1/metabolismo , Mesodermo/metabolismo , Microscopía Fluorescente , Persona de Mediana Edad , Fenotipo
12.
JCI Insight ; 4(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31581152

RESUMEN

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.


Asunto(s)
Tolerancia Inmunológica/fisiología , Trasplante de Islotes Pancreáticos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/fisiología , Supervivencia de Injerto , Humanos , Trasplante Homólogo
13.
Transplantation ; 102(10): e413-e423, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29975241

RESUMEN

BACKGROUND: There is a strong rationale to pursue the use of neonatal porcine islets (NPIs) as an unlimited source of islets for clinical xenotransplantation. Because NPIs are composed of immature insulin producing beta (ß) cells and ductal precursor cells, they provide an ideal model to examine culture conditions to enhance ß cell proliferation and/or ß cell neoformation from ductal cells. In an attempt to optimize the potential of NPIs as a source of ß cell grafts, we used an in vitro differentiation protocol and measured its effect on the functional maturation and differentiation of NPIs. METHODS: Pancreata from 1- to 3-day-old neonatal pigs were digested and cultured in standard Ham's F10 media for 5 days. Each independent preparation was then further cultured in Dulbecco's modified Eagle medium nutrient mixture-F12 differentiation media containing growth factors added in a stepwise fashion, or cultured in control Ham's F10 media. After 20 days in culture, islets were assessed for insulin secretory capacity, cellular composition, gene expression, and metabolic activity after transplantation in immunodeficient mice with diabetes. RESULTS: Compared with control islets, differentiated islets exhibited a significantly higher proportion of endocrine cells, proliferating cell nuclear antigen double positive ß cells, and an enhanced glucose-stimulated insulin secretory activity. Mice transplanted with differentiated islets had significantly lower blood glucose values at weeks 18 and 20 compared with nondifferentiated controls and were shown to be more glucose tolerant. CONCLUSIONS: Culturing NPIs in a 20-day stepwise differentiation media increases the proportion of endocrine cells and augments both in vitro and in vivo function of the islets.


Asunto(s)
Diabetes Mellitus Experimental/cirugía , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/fisiología , Técnicas de Cultivo de Tejidos/métodos , Trasplante Heterólogo/métodos , Animales , Animales Recién Nacidos , Glucemia , Diferenciación Celular , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Xenoinjertos/citología , Xenoinjertos/fisiología , Humanos , Insulina/metabolismo , Islotes Pancreáticos/citología , Masculino , Ratones , Estreptozocina/toxicidad , Sus scrofa , Resultado del Tratamiento
14.
Cell Death Dis ; 9(6): 595, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29789532

RESUMEN

Human islet transplantation has been hampered by donor cell death associated with the islet preparation procedure before transplantation. Regulated necrosis pathways are biochemically and morphologically distinct from apoptosis. Recently, ferroptosis was identified as a non-apoptotic form of iron-dependent regulated necrosis implicated in various pathological conditions. Mediators of islet oxidative stress, including glutathione peroxidase-4 (GPX4), have been identified as inhibitors of ferroptosis, and mechanisms that affect GPX4 function can impact islet function and viability. Ferroptosis has not been investigated directly in human islets, and its relevance in islet transplantation remains unknown. Herein, we sought to determine whether in vitro human islet viability and function is compromised in the presence of two distinct ferroptosis-inducing agents (FIA), erastin or RSL3, and whether these effects could be rescued with ferroptosis inhibitors, ferrostatin-1 (Fer-1), or desferrioxamine (DFO). Viability, as assessed by lactate dehydrogenase (LDH) release, revealed significant death in erastin- and RSL3-treated islets, 20.3% ± 3.8 and 24.4% ± 2.5, 24 h post culture, respectively. These effects were ameliorated in islets pre-treated with Fer-1 or the iron chelator, desferrioxamine (DFO). Stimulation index, a marker of islet function revealed a significant reduction in function in erastin-treated islets (control 1.97 ± 0.13 vs. 50 µM erastin 1.32 ± 0.1) (p < 0.05). Fer-1 and DFO pre-treatment alone did not augment islet viability or function. Pre-treatment of islets with erastin or Fer-1 did not impact in vivo engraftment in an immunodeficient mouse transplant model. Our data reveal that islets are indeed susceptible to ferroptosis in vitro, and induction of this novel cell death modality leads to compromised islet function, which can be recoverable in the presence of the ferroptosis inhibitors. The in vivo impact of this pathway in islet transplantation remains elusive given the constraints of our study, but warrants continued investigation.


Asunto(s)
Apoptosis , Hierro/metabolismo , Islotes Pancreáticos/fisiología , Supervivencia Tisular , Animales , Apoptosis/efectos de los fármacos , Carbolinas/farmacología , Células Cultivadas , Ciclohexilaminas/farmacología , Deferoxamina/farmacología , Glucosa/farmacología , Humanos , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Ratones Endogámicos C57BL , Fenilendiaminas/farmacología , Piperazinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Supervivencia Tisular/efectos de los fármacos , gamma-Glutamilciclotransferasa/genética , gamma-Glutamilciclotransferasa/metabolismo
15.
Diabetes ; 66(5): 1312-1321, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28246290

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Trasplante de Células Madre Mesenquimatosas , Neovascularización Fisiológica , Animales , Animales Recién Nacidos , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/irrigación sanguínea , Masculino , Ratones , Porcinos , Trasplantes/irrigación sanguínea
16.
Clin Cancer Res ; 8(2): 502-13, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11839670

RESUMEN

PURPOSE: The transformation status and role of clonotypic pre-switch IgM in the evolution of malignant post-switch multiple myeloma (MM) cells is unclear. In this study, we determined the differentiation stage within the B lineage of clonotypic cells from malignant and nonclinical isotype pools by analyzing the frequency and intraclonal diversity of members within each isotype pool. RESULTS: Immunoglobulin VDJ transcripts were amplified from peripheral blood cells of seven patients with a hemi-nested reverse transcription-PCR with complementarity determining region 1 (CDR1)-specific and constant region primers. Of the 1951 clones screened by patient CDR2/3-specific PCR, 356 of these were sequenced. Intraclonal homogeneity was observed in pre-switch transcripts from four of four informative patients. Transcripts from the IgM pool were relatively frequent in two of four informative patients. Cellular limiting dilution analysis indicated 0.4-25% of peripheral blood mononuclear cells expressed clonotypic IgM for 6 of 15 samples tested. By contrast, significant intraclonal diversity was observed in the nonclinical IgA pool of 1 patient. A genealogical tree of IgA sequences was constructed showing ongoing clonal diversification from sequences with close homology to the germ-line V gene to those resembling the PC sequence. Furthermore, some clones exhibited complete homology with tumor VDJ sequence, plus extra mutations, suggestive of a parallel clonal arm that remains responsive to an antigenic stimulus. CONCLUSIONS: Detection of intraclonal diversity in the post-switch nonclinical isotype pool suggests that remnants of the parent B-cell clone coexist with malignant clonal precursors. The presence of intraclonal homogeneity in the pre-switch IgM pool supports the idea that pre-switch MM cells play a role in malignant events within the MM clone.


Asunto(s)
Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Secuencia de Bases , Diferenciación Celular , Clonación Molecular , Reordenamiento Génico , Humanos , Cambio de Clase de Inmunoglobulina , Modelos Biológicos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Homología de Secuencia de Ácido Nucleico
17.
Exp Hematol ; 30(3): 221-8, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11882359

RESUMEN

OBJECTIVE: In multiple myeloma (MM), the immunoglobulin gene rearrangement characterizing malignant plasma cells is unique. For a patient with multiple myeloma who underwent a B-cell leukemic blast transformation, using the immunoglobulin molecular signature, we characterized the clonal relationship to autologous plasma cells and the impact on normal polyclonal B-lymphocyte populations. METHODS: Single-cell reverse transcriptase polymerase chain reaction (RT-PCR)/PCR was used to determine the clonal relationship between autologous MM plasma cells and leukemic B cells. A murine xenograft model was used to determine the myelomagenic potential of the leukemic B cells. RESULTS: Single-cell analysis showed that circulating leukemic-phase cells were clonotypic, with an IgH VDJ sequence identical to that of diagnosis plasma cells. Analysis of IgH transcripts indicates MM clonal dominance over normal B-cell components of the immune system at diagnosis and during leukemic disease. Leukemic B cells were xenografted to irradiated NOD/SCID mice, leading to lytic bone lesions and clonotypic cells in murine BM. Although human cells in murine BM expressed CD138, a marker largely absent from ex vivo leukemic cells, the expression of CD45, CD19, and CD20 confirmed that engrafting cells were mature, probably late-stage B cells rather than plasma cells. CONCLUSIONS: Leukemic B cells are able to exert strong clonal dominance over normal components of the immune system, colonize the murine BM in a xenograft model, and disrupt normal bone metabolism leading to lytic bone lesions. This supports the hypothesis that clonotypic MM B cells are reservoirs of disease that persist throughout therapy and give rise to relapse.


Asunto(s)
Médula Ósea/patología , Células Clonales/patología , Leucemia de Células B/genética , Mieloma Múltiple/genética , Animales , Antígenos CD19/análisis , Antígenos CD20/análisis , Linfocitos B/inmunología , Linfocitos B/patología , Reordenamiento Génico de Cadena Pesada de Linfocito B , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Leucemia de Células B/inmunología , Leucemia de Células B/patología , Antígenos Comunes de Leucocito/análisis , Activación de Linfocitos , Masculino , Glicoproteínas de Membrana/análisis , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Trasplante de Neoplasias , Células Plasmáticas/patología , Proteoglicanos/análisis , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sindecano-1 , Sindecanos , Trasplante Heterólogo
18.
World J Diabetes ; 5(1): 59-68, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24567802

RESUMEN

AIM: To minimize the expansion of pancreatic mesenchymal cells in vitro and confirm that ß-cell progenitors reside within the pancreatic epithelium. METHODS: Due to mesenchymal stem cell (MSC) expansion and overgrowth, progenitor cells within the pancreatic epithelium cannot be characterized in vitro, though ß-cell dedifferentiation and expansion of MSC intermediates via epithelial-mesenchymal transition (EMT) may generate ß-cell progenitors. Pancreatic epithelial cells from endocrine and non-endocrine tissue were expanded and differentiated in a novel pancreatic epithelial expansion medium supplemented with growth factors known to support epithelial cell growth (dexamethasone, epidermal growth factor, 3,5,3'-triiodo-l-thyronine, bovine brain extract). Cells were also infected with a single and dual lentiviral reporter prior to cell differentiation. Enhanced green fluorescent protein was controlled by the rat Insulin 1 promoter and the monomeric red fluorescent protein was controlled by the mouse PDX1 promoter. In combination with lentiviral tracing, cells expanded and differentiated in the pancreatic medium were characterized by flow cytometry (BD fluorescence activated cell sorting), immunostaining and real-time polymerase chain reaction (PCR) (7900HT Fast Realtime PCR System). RESULTS: In the presence of 10% serum MSCs rapidly expand in vitro while the epithelial cell population declines. The percentage of vimentin(+) cells increased from 22% ± 5.83% to 80.43% ± 3.24% (14 d) and 99.00% ± 0.0% (21 d), and the percentage of epithelial cells decreased from 74.71% ± 8.34% to 26.57% ± 9.75% (14 d) and 4.00% ± 1.53% (21 d), P < 0.01 for all time points. Our novel pancreatic epithelial expansion medium preserved the epithelial cell phenotype and minimized epithelial cell dedifferentiation and EMT. Cells expanded in our epithelial medium contained significantly less mesenchymal cells (vimentin(+)) compared to controls (44.87% ± 4.93% vs 95.67% ± 1.36%; P < 0.01). During cell differentiation lentiviral reporting demonstrated that, PDX1(+) and insulin(+) cells were localized within adherent epithelial cell aggregates compared to controls. Compared to starting islets differentiated cells had at least two fold higher gene expression of PDX1, insulin, PAX4 and RFX (P < 0.05). CONCLUSION: PDX1(+) cells were confined to adherent epithelial cell aggregates and not vimentin(+) cells (mesenchymal), suggesting that EMT is not a mechanism for generating pancreatic progenitor cells.

19.
Islets ; 5(5): 216-25, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24262950

RESUMEN

Islet transplantation is a promising treatment for Type 1 diabetes; however limitations of the intra-portal site and poor revascularization of islets must be overcome. We hypothesize that engineering a highly vascularized collagen-based construct will allow islet graft survival and function in alternative sites. In this study, we developed such a collagen-based biomaterial. Neonatal porcine islets (NPIs) were embedded in collagen matrices crosslinked with 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide and N-hydroxysuccinimide containing combinations of chondroitin-6-sulfate, chitosan, and laminin, and compared with controls cultured in standard media. Islets were examined for insulin secretory activity after 24 h and 4 d and for apoptotic cell death and matrix integrity after 7 d in vitro. These same NPI/collagen constructs were transplanted subcutaneously in immunoincompetent B6.Rag-/- mice and then assessed for islet survival and vascularization. At all time points assessed during in vitro culture there were no significant differences in insulin secretory activity between control islets and those embedded in the collagen constructs, indicating that the collagen matrix had no adverse effect on islet function. Less cell death was observed in the matrix with all co-polymers compared with the other matrices tested. Immunohistochemical analysis of the grafts post-transplant confirmed the presence of intact insulin-positive islets; grafts were also shown to be vascularized by von Willebrand factor staining. This study demonstrates that a collagen, chondroitin-6-sulfate, chitosan, and laminin matrix supports islet function in vitro and moreover allows islet survival and vascularization post-transplantation; therefore, this bio-engineered vascularized construct is capable of supporting islet survival.


Asunto(s)
Colágeno , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Islotes Pancreáticos/fisiología , Islotes Pancreáticos/irrigación sanguínea , Técnicas de Cultivo de Tejidos/métodos , Trasplante Heterotópico/métodos , Animales , Apoptosis , Bioingeniería , Carbodiimidas , Quitosano , Sulfatos de Condroitina , Reactivos de Enlaces Cruzados , Medios de Cultivo/química , Supervivencia de Injerto , Insulina/análisis , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/química , Islotes Pancreáticos/metabolismo , Laminina , Ratones , Ratones Endogámicos BALB C , Neovascularización Fisiológica , Succinimidas , Porcinos
20.
Endocrinology ; 154(4): 1392-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23449893

RESUMEN

Our understanding of adult human ß-cells is advancing, but we know little about the function and plasticity of ß-cells from infants. We therefore characterized islets and single islet cells from human infants after isolation and culture. Although islet morphology in pancreas biopsies was similar to that in adults, infant islets after isolation and 24-48 hours of culture had less insulin staining, content, and secretion. The cultured infant islets expressed pancreatic and duodenal homeobox 1 and several (Glut1, Cav1.3, Kir6.2) but not all (syntaxin 1A and synaptosomal-associated protein 25) markers of functional islets, suggesting a loss of secretory phenotype in culture. The activity of key ion channels was maintained in isolated infant ß-cells, whereas exocytosis was much lower than in adults. We examined whether a functional exocytotic phenotype could be reestablished under conditions thought to promote ß-cell differentiation. After a 24- to 28-day expansion and maturation protocol, we found preservation of endocrine markers and hormone expression, an increased proportion of insulin-positive cells, elevated expression of syntaxin 1A and synaptosomal-associated protein 25, and restoration of exocytosis to levels comparable with that in adult ß-cells. Thus, human infant islets are prone to loss of their exocytotic phenotype in culture but amenable to experimental approaches aimed at promoting expansion and functional maturation. Control of exocytotic protein expression may be an important mechanism underlying the plasticity of the secretory machinery, an increased understanding of which may lead to improved regenerative approaches to treat diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Canales de Calcio Tipo L/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Exocitosis/fisiología , Femenino , Glucagón/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Lactante , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/crecimiento & desarrollo , Masculino , Persona de Mediana Edad , Técnicas de Placa-Clamp , Fenotipo , Canales de Potasio de Rectificación Interna/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sintaxina 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA