Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Transpl Int ; 36: 11367, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37359825

RESUMEN

Long-term success in beta-cell replacement remains limited by the toxic effects of calcineurin inhibitors (CNI) on beta-cells and renal function. We report a multi-modal approach including islet and pancreas-after-islet (PAI) transplant utilizing calcineurin-sparing immunosuppression. Ten consecutive non-uremic patients with Type 1 diabetes underwent islet transplant with immunosuppression based on belatacept (BELA; n = 5) or efalizumab (EFA; n = 5). Following islet failure, patients were considered for repeat islet infusion and/or PAI transplant. 70% of patients (four EFA, three BELA) maintained insulin independence at 10 years post-islet transplant, including four patients receiving a single islet infusion and three patients undergoing PAI transplant. 60% remain insulin independent at mean follow-up of 13.3 ± 1.1 years, including one patient 9 years after discontinuing all immunosuppression for adverse events, suggesting operational tolerance. All patients who underwent repeat islet transplant experienced graft failure. Overall, patients demonstrated preserved renal function, with a mild decrease in GFR from 76.5 ± 23.1 mL/min to 50.2 ± 27.1 mL/min (p = 0.192). Patients undergoing PAI showed the greatest degree of renal impairment following initiation of CNI (56% ± 18.7% decrease in GFR). In our series, repeat islet transplant is ineffective at maintaining long-term insulin independence. PAI results in durable insulin independence but is associated with impaired renal function secondary to CNI dependence.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Islotes Pancreáticos , Trasplante de Páncreas , Humanos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/cirugía , Insulina/uso terapéutico , Calcineurina , Terapia de Inmunosupresión/métodos , Trasplante de Islotes Pancreáticos/métodos , Inhibidores de la Calcineurina/uso terapéutico , Inmunosupresores/uso terapéutico
2.
Gut ; 71(9): 1831-1842, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34702715

RESUMEN

OBJECTIVE: Chronic pancreatitis (CP) is a potentially fatal disease of the exocrine pancreas, with no specific or effective approved therapies. Due to difficulty in accessing pancreas tissues, little is known about local immune responses or pathogenesis in human CP. We sought to characterise pancreatic immune responses using tissues derived from patients with different aetiologies of CP and non-CP organ donors in order to identify key signalling molecules associated with human CP. DESIGN: We performed single-cell level cellular indexing of transcriptomes and epitopes by sequencing and T-cell receptor (TCR) sequencing of pancreatic immune cells isolated from organ donors, hereditary and idiopathic patients with CP who underwent total pancreatectomy. We validated gene expression data by performing flow cytometry and functional assays in a second patient with CP cohort. RESULTS: Deep single-cell sequencing revealed distinct immune characteristics and significantly enriched CCR6+ CD4+ T cells in hereditary compared with idiopathic CP. In hereditary CP, a reduction in T-cell clonality was observed due to the increased CD4+ T (Th) cells that replaced tissue-resident CD8+ T cells. Shared TCR clonotype analysis among T-cell lineages also unveiled unique interactions between CCR6+ Th and Th1 subsets, and TCR clustering analysis showed unique common antigen binding motifs in hereditary CP. In addition, we observed a significant upregulation of the CCR6 ligand (CCL20) expression among monocytes in hereditary CP as compared with those in idiopathic CP. The functional significance of CCR6 expression in CD4+ T cells was confirmed by flow cytometry and chemotaxis assay. CONCLUSION: Single-cell sequencing with pancreatic immune cells in human CP highlights pancreas-specific immune crosstalk through the CCR6-CCL20 axis, a signalling pathway that might be leveraged as a potential future target in human hereditary CP.


Asunto(s)
Pancreatitis Crónica , Receptores CCR6 , Inmunidad Adaptativa , Linfocitos T CD8-positivos , Quimiocina CCL20/metabolismo , Citometría de Flujo , Humanos , Pancreatitis Crónica/genética , Receptores CCR6/genética , Receptores CCR6/metabolismo
3.
Am J Transplant ; 20(8): 2091-2100, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31994295

RESUMEN

The approach to transplantation in human immunodeficiency virus (HIV)-positive patients has been conservative due to fear of exacerbating an immunocompromised condition. As a result, HIV-positive patients with diabetes were initially excluded from beta cell replacement therapy. Early reports of pancreas transplant in patients with HIV described high rates of early graft loss with limited follow-up. We report long-term follow-up of islet or pancreas transplantation in HIV-positive type 1 diabetic patients who received a kidney transplant concurrently or had previously undergone kidney transplantation. Although 4 patients developed polyoma viremia, highly active antiretroviral therapy and adequate infectious prophylaxis were successful in providing protection until CD4+ counts recovered. Coordination with HIV providers is critical to reduce the risk of rejection by minimizing drug-drug interactions. Also, protocols for prophylaxis of opportunistic infections and strategies for monitoring and treating BK viremia are important given the degree of immunosuppression required. This series demonstrates that type 1 diabetic patients with well-controlled HIV and renal failure can be appropriate candidates for beta cell replacement, with a low rate of infectious complications, early graft loss, and rejection, so excellent long-term graft survival is possible. Additionally, patients with HIV and cardiovascular contraindications can undergo islet infusion.


Asunto(s)
Diabetes Mellitus Tipo 1 , Infecciones por VIH , Seropositividad para VIH , Trasplante de Páncreas , Insuficiencia Renal , Diabetes Mellitus Tipo 1/complicaciones , Estudios de Seguimiento , Rechazo de Injerto/etiología , Supervivencia de Injerto , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Humanos , Trasplante de Páncreas/efectos adversos
4.
Transpl Int ; 33(11): 1516-1528, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32852857

RESUMEN

The success of pancreas islet isolation largely depends on donor characteristics, including extracellular matrix composition of which collagen is the main element. We hypothesized that isolation yields are proportional to collagen digestion percentage, and aimed to determine a threshold that predicts isolation success. The amount of pancreas collagen (I-V) was determined using colorimetry prior to and after the digestion process in 52 human islet isolations. Collagen I-V and VI were also assessed histologically. We identified a collagen digestion threshold of ≥ 60% as an independent factor beyond which an islet preparation has a ninefold increased odds of yielding ≥ 250 000 islet equivalents (IEQ) (P = 0.009) and a sixfold increased odds of being transplanted (P = 0.015). Preparations with ≥ 60% collagen digestion (n = 35) yielded 283 017 ± 164 214 IEQ versus 180 142 ± 85 397 in the < 60% collagen digestion group (n = 17) (P = 0.016); respectively 62.9% versus 29.4% of those were transplanted (P = 0.024). Common donor characteristics, initial collagen content, enzyme blend, and digestion times were not associated with collagen digestion percentage variations. Donor age positively correlated with the amount of collagen VI (P = 0.013). There was no difference in islet graft survival between high and low digestion groups. We determined that a 60% pancreas collagen digestion is the threshold beyond which an islet isolation is likely to be successful and transplanted.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Separación Celular , Colágeno , Digestión , Humanos , Páncreas , Estudios Prospectivos
5.
Nature ; 455(7216): 1109-13, 2008 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-18806780

RESUMEN

Type 1 diabetes (T1D) is a debilitating autoimmune disease that results from T-cell-mediated destruction of insulin-producing beta-cells. Its incidence has increased during the past several decades in developed countries, suggesting that changes in the environment (including the human microbial environment) may influence disease pathogenesis. The incidence of spontaneous T1D in non-obese diabetic (NOD) mice can be affected by the microbial environment in the animal housing facility or by exposure to microbial stimuli, such as injection with mycobacteria or various microbial products. Here we show that specific pathogen-free NOD mice lacking MyD88 protein (an adaptor for multiple innate immune receptors that recognize microbial stimuli) do not develop T1D. The effect is dependent on commensal microbes because germ-free MyD88-negative NOD mice develop robust diabetes, whereas colonization of these germ-free MyD88-negative NOD mice with a defined microbial consortium (representing bacterial phyla normally present in human gut) attenuates T1D. We also find that MyD88 deficiency changes the composition of the distal gut microbiota, and that exposure to the microbiota of specific pathogen-free MyD88-negative NOD donors attenuates T1D in germ-free NOD recipients. Together, these findings indicate that interaction of the intestinal microbes with the innate immune system is a critical epigenetic factor modifying T1D predisposition.


Asunto(s)
Bacterias/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Inmunidad Innata/inmunología , Intestinos/microbiología , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/genética , Femenino , Inmunidad Innata/genética , Interferón gamma/inmunología , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Datos de Secuencia Molecular , Factor 88 de Diferenciación Mieloide/genética , Filogenia , Organismos Libres de Patógenos Específicos , Factores de Tiempo
6.
Bioeng Transl Med ; 8(2): e10444, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36925691

RESUMEN

Clinical islet transplantation for treatment of type 1 diabetes (T1D) is limited by the shortage of pancreas donors and need for lifelong immunosuppressive therapy. A convection-driven intravascular bioartificial pancreas (iBAP) based on highly permeable, yet immunologically protective, silicon nanopore membranes (SNM) holds promise to sustain islet function without the need for immunosuppressants. Here, we investigate short-term functionality of encapsulated human islets in an iBAP prototype. Using the finite element method (FEM), we calculated predicted oxygen profiles within islet scaffolds at normalized perifusion rates of 14-200 nl/min/IEQ. The modeling showed the need for minimum in vitro and in vivo islet perifusion rates of 28 and 100 nl/min/IEQ, respectively to support metabolic insulin production requirements in the iBAP. In vitro glucose-stimulated insulin secretion (GSIS) profiles revealed a first-phase response time of <15 min and comparable insulin production rates to standard perifusion systems (~10 pg/min/IEQ) for perifusion rates of 100-200 nl/min/IEQ. An intravenous glucose tolerance test (IVGTT), performed at a perifusion rate of 100-170 nl/min/IEQ in a non-diabetic pig, demonstrated a clinically relevant C-peptide production rate (1.0-2.8 pg/min/IEQ) with a response time of <5 min.

7.
J Exp Med ; 203(7): 1701-11, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16818678

RESUMEN

Regulatory T (T reg) cells are critical regulators of immune tolerance. Most T reg cells are defined based on expression of CD4, CD25, and the transcription factor, FoxP3. However, these markers have proven problematic for uniquely defining this specialized T cell subset in humans. We found that the IL-7 receptor (CD127) is down-regulated on a subset of CD4(+) T cells in peripheral blood. We demonstrate that the majority of these cells are FoxP3(+), including those that express low levels or no CD25. A combination of CD4, CD25, and CD127 resulted in a highly purified population of T reg cells accounting for significantly more cells that previously identified based on other cell surface markers. These cells were highly suppressive in functional suppressor assays. In fact, cells separated based solely on CD4 and CD127 expression were anergic and, although representing at least three times the number of cells (including both CD25(+)CD4(+) and CD25(-)CD4(+) T cell subsets), were as suppressive as the "classic" CD4(+)CD25(hi) T reg cell subset. Finally, we show that CD127 can be used to quantitate T reg cell subsets in individuals with type 1 diabetes supporting the use of CD127 as a biomarker for human T reg cells.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Receptores de Interleucina-7/biosíntesis , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Biomarcadores , Recuento de Linfocito CD4 , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Factores de Transcripción Forkhead/genética , Humanos , Memoria Inmunológica , Persona de Mediana Edad , Receptores de Interleucina-2/biosíntesis , Receptores de Interleucina-2/genética , Receptores de Interleucina-7/genética , Linfocitos T Reguladores/metabolismo
8.
J Immunol ; 182(5): 2590-600, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19234153

RESUMEN

Dendritic cells (DCs) are essential in T cell-mediated destruction of insulin-producing beta cells in the islets of Langerhans in type 1 diabetes. In this study, we investigated T cell induction of intra-islet DC maturation during the progression of the disease in both autoimmune-prone NOD and resistant C57BL/6 mice. We demonstrated steady-state capture and retention of unprocessed beta cell-derived proteins by semimature intra-islet DCs in both mouse strains. T cell-mediated intra-islet inflammation induced an increase in CD40 and CD80 expression and processing of captured Ag by resident DCs without inducing the expression of the p40 subunit of IL-12/23. Some of the CD40(high) intra-islet DCs up-regulated CCR7, and a small number of CD40(high) DCs bearing unprocessed islet Ags were detected in the pancreatic lymph nodes in mice with acute intra-islet inflammation, demonstrating that T cell-mediated tissue inflammation augments migration of mature resident DCs to draining lymph nodes. Our results identify an amplification loop during the progression of autoimmune diabetes, in which initial T cell infiltration leads to rapid maturation of intra-islet DCs, their migration to lymph nodes, and expanded priming of more autoreactive T cells. Therapeutic interventions that intercept this process may be effective at halting the progression of type 1 diabetes.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/patología , Animales , Enfermedades Autoinmunes/metabolismo , Diferenciación Celular/genética , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas de Homeodominio/fisiología , Inmunofenotipificación , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Linfocitos T/inmunología , Transactivadores/fisiología
9.
Proc Natl Acad Sci U S A ; 105(48): 18895-900, 2008 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19015530

RESUMEN

The recent development of small-molecule tyrosine kinase (TK) inhibitors offers increasing opportunities for the treatment of autoimmune diseases. In this study, we investigated the potential of this new class of drugs to treat and cure type 1 diabetes (T1D) in the NOD mouse. Treatment of prediabetic and new onset diabetic mice with imatinib (Gleevec) prevented and reversed T1D. Similar results were observed with sunitinib (Sutent), an additional approved multikinase inhibitor, suggesting that the primary target of imatinib, c-Abl, was not essential in blocking disease in this model. Additional studies with another TK inhibitor, PLX647 (targeting c-Kit and c-Fms) or an anti-c-Kit mAb showed only marginal efficacy whereas a soluble form of platelet-derived growth factor receptor (PDGFR), PDGFRbetaIg, rapidly reversed diabetes. These findings strongly suggest that inhibition of PDGFR is critical to reverse diabetes and highlight a crucial role of inflammation in the development of T1D. These conclusions were supported by the finding that the adaptive immune system was not significantly affected by imatinib treatment. Finally, and most significantly, imatinib treatment led to durable remission after discontinuation of therapy at 10 weeks in a majority of mice. Thus, long-term efficacy and tolerance is likely to depend on inhibiting a combination of tyrosine kinases supporting the use of selective kinase inhibitors as a new, potentially very attractive approach for the treatment of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Ratones Endogámicos NOD/metabolismo , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Benzamidas , Glucemia/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/veterinaria , Femenino , Humanos , Mesilato de Imatinib , Indoles/uso terapéutico , Leucocitos/metabolismo , Ratones , Ratones SCID , Páncreas/citología , Páncreas/patología , Proteínas Tirosina Quinasas/metabolismo , Pirroles/uso terapéutico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inducción de Remisión , Sunitinib
10.
J Biomed Mater Res A ; 109(12): 2438-2448, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34196100

RESUMEN

Type 1 diabetic patients with severe hypoglycemia unawareness have benefitted from cellular therapies, such as pancreas or islet transplantation; however, donor shortage and the need for immunosuppression limits widespread clinical application. We previously developed an intravascular bioartificial pancreas (iBAP) using silicon nanopore membranes (SNM) for immunoprotection. To ensure ample nutrient delivery, the iBAP will need a cell scaffold with high hydraulic permeability to provide mechanical support and maintain islet viability and function. Here, we examine the feasibility of superporous agarose (SPA) as a potential cell scaffold in the iBAP. SPA exhibits 66-fold greater hydraulic permeability than the SNM along with a short (<10 µm) diffusion distance to the nearest islet. SPA also supports short-term functionality of both encapsulated human islets and stem-cell-derived enriched ß-clusters in a convection-based system, demonstrated by high viability (>95%) and biphasic insulin responses to dynamic glucose stimulus. These findings suggest that the SPA scaffold will not limit nutrient delivery in a convection-based bioartificial pancreas and merits continued investigation.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Páncreas Artificial , Sefarosa/química , Trasplante de Células Madre/métodos , Andamios del Tejido , Adulto , Diabetes Mellitus Tipo 1/terapia , Glucosa/farmacología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Membranas Artificiales , Nanoporos , Silicio
11.
J Clin Med ; 10(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209541

RESUMEN

The Food and Drug Administration (FDA) has been regulating human islets for allotransplantation as a biologic drug in the US. Consequently, the requirement of a biological license application (BLA) approval before clinical use of islet transplantation as a standard of care procedure has stalled the development of the field for the last 20 years. Herein, we provide our commentary to the multiple FDA's position papers and guidance for industry arguing that BLA requirement has been inappropriately applied to allogeneic islets, which was delivered to the FDA Cellular, Tissue and Gene Therapies Advisory Committee on 15 April 2021. We provided evidence that BLA requirement and drug related regulations are inadequate in reassuring islet product quality and potency as well as patient safety and clinical outcomes. As leaders in the field of transplantation and endocrinology under the "Islets for US Collaborative" designation, we examined the current regulatory status of islet transplantation in the US and identified several anticipated negative consequences of the BLA approval. In our commentary we also offer an alternative pathway for islet transplantation under the regulatory framework for organ transplantation, which would address deficiencies of in current system.

12.
J Clin Invest ; 130(5): 2705-2711, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32053120

RESUMEN

Chronic pancreatitis (CP) is considered an irreversible fibroinflammatory pancreatic disease. Despite numerous animal model studies, questions remain about local immune characteristics in human CP. We profiled pancreatic immune cell characteristics in control organ donors and CP patients including those with hereditary and idiopathic CP undergoing total pancreatectomy with islet autotransplantation. Flow cytometric analysis revealed a significant increase in the frequency of CD68+ macrophages in idiopathic CP. In contrast, hereditary CP samples showed a significant increase in CD3+ T cell frequency, which prompted us to investigate the T cell receptor ß (TCRß) repertoire in the CP and control groups. TCRß sequencing revealed a significant increase in TCRß repertoire diversity and reduced clonality in both CP groups versus controls. Interestingly, we observed differences in Vß-Jß gene family usage between hereditary and idiopathic CP and a positive correlation of TCRß rearrangements with disease severity scores. Immunophenotyping analyses in hereditary and idiopathic CP pancreases indicate differences in innate and adaptive immune responses, which highlights differences in immunopathogenic mechanisms of disease among subtypes of CP. TCR repertoire analysis further suggests a role for specific T cell responses in hereditary versus idiopathic CP pathogenesis, providing insights into immune responses associated with human CP.


Asunto(s)
Pancreatitis Crónica/genética , Pancreatitis Crónica/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Femenino , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Humanos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Páncreas/inmunología , Páncreas/patología , Pancreatitis Crónica/patología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología
13.
Nat Cell Biol ; 21(6): 792, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30914825

RESUMEN

In the version of this article originally published, the Gene Expression Omnibus (GEO) accession number listed in the data availability section was incorrectly given as GSE10979 instead of GSE109795. The sentence should read "RNA-seq data that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE109795," and the code should link to https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE109795. The error has been corrected in the HTML and PDF versions of the paper.

14.
Nat Cell Biol ; 21(2): 263-274, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30710150

RESUMEN

Despite advances in the differentiation of insulin-producing cells from human embryonic stem cells, the generation of mature functional ß cells in vitro has remained elusive. To accomplish this goal, we have developed cell culture conditions to closely mimic events occurring during pancreatic islet organogenesis and ß cell maturation. In particular, we have focused on recapitulating endocrine cell clustering by isolating and reaggregating immature ß-like cells to form islet-sized enriched ß-clusters (eBCs). eBCs display physiological properties analogous to primary human ß cells, including robust dynamic insulin secretion, increased calcium signalling in response to secretagogues, and improved mitochondrial energization. Notably, endocrine cell clustering induces metabolic maturation by driving mitochondrial oxidative respiration, a process central to stimulus-secretion coupling in mature ß cells. eBCs display glucose-stimulated insulin secretion as early as three days after transplantation in mice. In summary, replicating aspects of endocrine cell clustering permits the generation of stem-cell-derived ß cells that resemble their endogenous counterparts.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Endocrinas/citología , Fibroblastos/citología , Células Madre Embrionarias Humanas/citología , Células Secretoras de Insulina/citología , Animales , Células Cultivadas , Células Madre Embrionarias/fisiología , Células Endocrinas/fisiología , Fibroblastos/fisiología , Glucosa/farmacología , Células Madre Embrionarias Humanas/fisiología , Humanos , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/citología , Ratones , Mitocondrias/metabolismo
15.
Transplantation ; 103(1): 160-167, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30095738

RESUMEN

BACKGROUND: All human islets used in research and for the clinical treatment of diabetes are subject to ischemic damage during pancreas procurement, preservation, and islet isolation. A major factor influencing islet function is exposure of pancreata to cold ischemia during unavoidable windows of preservation by static cold storage (SCS). Improved preservation methods may prevent this functional deterioration. In the present study, we investigated whether pancreas preservation by gaseous oxygen perfusion (persufflation) better preserved islet function versus SCS. METHODS: Human pancreata were preserved by SCS or by persufflation in combination with SCS. Islets were subsequently isolated, and preparations in each group matched for SCS or total preservation time were compared using dynamic glucose-stimulated insulin secretion as a measure of ß-cell function and RNA sequencing to elucidate transcriptomic changes. RESULTS: Persufflated pancreata had reduced SCS time, which resulted in islets with higher glucose-stimulated insulin secretion compared to islets from SCS only pancreata. RNA sequencing of islets from persufflated pancreata identified reduced inflammatory and greater metabolic gene expression, consistent with expectations of reducing cold ischemic exposure. Portions of these transcriptional responses were not associated with time spent in SCS and were attributable to pancreatic reoxygenation. Furthermore, persufflation extended the total preservation time by 50% without any detectable decline in islet function or viability. CONCLUSIONS: These data demonstrate that pancreas preservation by persufflation rather than SCS before islet isolation reduces inflammatory responses and promotes metabolic pathways in human islets, which results in improved ß cell function.


Asunto(s)
Frío , Mediadores de Inflamación/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Preservación de Órganos/métodos , Oxígeno/farmacología , Perfusión/métodos , Adolescente , Adulto , Supervivencia Celular/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Persona de Mediana Edad , Preservación de Órganos/efectos adversos , Vías Secretoras/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Recolección de Tejidos y Órganos , Adulto Joven
16.
J Clin Invest ; 114(7): 979-87, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15467837

RESUMEN

The development of autoimmune diabetes in the nonobese diabetic (NOD) mouse results from a breakdown in tolerance to pancreatic islet antigens. CD28-B7 and CD40 ligand-CD40 (CD40L-CD40) costimulatory pathways affect the development of disease and are promising therapeutic targets. Indeed, it was shown previously that diabetes fails to develop in NOD-B7-2-/- and NOD-CD40L-/- mice. In this study, we examined the relative role of these 2 costimulatory pathways in the balance of autoimmunity versus regulation in NOD mice. We demonstrate that initiation but not effector function of autoreactive T cells was defective in NOD-B7-2-/- mice. Moreover, the residual proliferation of the autoreactive cells was effectively controlled by CD28-dependent CD4+CD25+ regulatory T cells (Treg's), as depletion of Treg's partially restored proliferation of autoreactive T cells and resulted in diabetes in an adoptive-transfer model. Similarly, disruption of the CD28-B7 pathway and subsequent Treg deletion restored autoimmunity in NOD-CD40L-/- mice. These results demonstrate that development of diabetes is dependent on a balance of pathogenic and regulatory T cells that is controlled by costimulatory signals. Thus, elimination of Treg's results in diabetes even in the absence of costimulation, which suggests a need for alternative strategies for immunotherapeutic approaches.


Asunto(s)
Antígenos CD28/metabolismo , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Subgrupos de Linfocitos T/fisiología , Linfocitos T/fisiología , Abatacept , Traslado Adoptivo , Animales , Glucemia/metabolismo , Antígenos CD28/genética , Antígenos CD40/genética , Ligando de CD40/genética , División Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Inmunoconjugados/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología
17.
Stem Cell Reports ; 9(3): 807-819, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28803916

RESUMEN

The advent of large-scale in vitro differentiation of human stem cell-derived insulin-producing cells (SCIPC) has brought us closer to treating diabetes using stem cell technology. However, decades of experiences from islet transplantation show that ischemia-induced islet cell death after transplant severely limits the efficacy of the therapy. It is unclear to what extent human SCIPC are susceptible to ischemia. In this study, we show that more than half of SCIPC die shortly after transplantation. Nutrient deprivation and hypoxia acted synergistically to kill SCIPC in vitro. Amino acid supplementation rescued SCIPC from nutrient deprivation, likely by providing cellular energy. Generating SCIPC under physiological oxygen tension of 5% conferred hypoxia resistance without affecting their differentiation or function. A two-pronged strategy of physiological oxygen acclimatization during differentiation and amino acid supplementation during transplantation significantly improved SCIPC survival after transplant.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Isquemia/terapia , Trasplante de Islotes Pancreáticos , Trasplante de Células Madre , Células Madre/metabolismo , Aminoácidos/farmacología , Animales , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Citoprotección/efectos de los fármacos , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Isquemia/patología , Ratones Endogámicos C57BL , Oxígeno/farmacología , Ácido Pirúvico/farmacología , Células Madre/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Supervivencia Tisular/efectos de los fármacos
18.
Nat Commun ; 8: 14686, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28272465

RESUMEN

Development of systems that reconstitute hallmark features of human pancreatic intraepithelial neoplasia (PanINs), the precursor to pancreatic ductal adenocarcinoma, could generate new strategies for early diagnosis and intervention. However, human cell-based PanIN models with defined mutations are unavailable. Here, we report that genetic modification of primary human pancreatic cells leads to development of lesions resembling native human PanINs. Primary human pancreas duct cells harbouring oncogenic KRAS and induced mutations in CDKN2A, SMAD4 and TP53 expand in vitro as epithelial spheres. After pancreatic transplantation, mutant clones form lesions histologically similar to native PanINs, including prominent stromal responses. Gene expression profiling reveals molecular similarities of mutant clones with native PanINs, and identifies potential PanIN biomarker candidates including Neuromedin U, a circulating peptide hormone. Prospective reconstitution of human PanIN development from primary cells provides experimental opportunities to investigate pancreas cancer development, progression and early-stage detection.


Asunto(s)
Adenocarcinoma in Situ/genética , Carcinoma Ductal Pancreático/genética , Conductos Pancreáticos/citología , Neoplasias Pancreáticas/genética , Adulto , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular , Trasplante de Células , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mutación , Neuropéptidos/metabolismo , Conductos Pancreáticos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína Smad4/genética , Transcriptoma , Proteína p53 Supresora de Tumor/genética
19.
Diabetes ; 65(11): 3418-3428, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27465220

RESUMEN

Eight manufacturing facilities participating in the National Institutes of Health-sponsored Clinical Islet Transplantation (CIT) Consortium jointly developed and implemented a harmonized process for the manufacture of allogeneic purified human pancreatic islet (PHPI) product evaluated in a phase 3 trial in subjects with type 1 diabetes. Manufacturing was controlled by a common master production batch record, standard operating procedures that included acceptance criteria for deceased donor organ pancreata and critical raw materials, PHPI product specifications, certificate of analysis, and test methods. The process was compliant with Current Good Manufacturing Practices and Current Good Tissue Practices. This report describes the manufacturing process for 75 PHPI clinical lots and summarizes the results, including lot release. The results demonstrate the feasibility of implementing a harmonized process at multiple facilities for the manufacture of a complex cellular product. The quality systems and regulatory and operational strategies developed by the CIT Consortium yielded product lots that met the prespecified characteristics of safety, purity, potency, and identity and were successfully transplanted into 48 subjects. No adverse events attributable to the product and no cases of primary nonfunction were observed.


Asunto(s)
Trasplante de Islotes Pancreáticos/métodos , Adolescente , Adulto , Anciano , Femenino , Humanos , Islotes Pancreáticos , Trasplante de Islotes Pancreáticos/economía , Masculino , Persona de Mediana Edad , National Institutes of Health (U.S.) , Estados Unidos , Adulto Joven
20.
Cell Transplant ; 25(8): 1515-1523, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26922947

RESUMEN

Selection of an optimal donor pancreas is the first key task for successful islet isolation. We conducted a retrospective multicenter study in 11 centers in North America to develop an islet donor scoring system using donor variables. The data set consisting of 1,056 deceased donors was used for development of a scoring system to predict islet isolation success (defined as postpurification islet yield >400,000 islet equivalents). With the aid of univariate logistic regression analyses, we developed the North American Islet Donor Score (NAIDS) ranging from 0 to 100 points. The c index in the development cohort was 0.73 (95% confidence interval 0.70-0.76). The success rate increased proportionally as the NAIDS increased, from 6.8% success in the NAIDS < 50 points to 53.7% success in the NAIDS ≥ 80 points. We further validated the NAIDS using a separate set of data consisting of 179 islet isolations. A comparable outcome of the NAIDS was observed in the validation cohort. The NAIDS may be a useful tool for donor pancreas selection in clinical practice. Apart from its utility in clinical decision making, the NAIDS may also be used in a research setting as a standardized measurement of pancreas quality.


Asunto(s)
Trasplante de Islotes Pancreáticos/métodos , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Páncreas/cirugía , Estudios Retrospectivos , Donantes de Tejidos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA