Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.536
Filtrar
1.
J Cell Sci ; 137(20)2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38804679

RESUMEN

The definitive demonstration of protein localization on primary cilia has been a challenge for cilia biologists. Primary cilia are solitary thread-like projections that have a specialized protein composition, but as the ciliary structure overlays the cell membrane and other cell parts, the identity of ciliary proteins are difficult to ascertain by conventional imaging approaches like immunofluorescence microscopy. Surface scanning electron microscopy combined with immunolabeling (immuno-SEM) bypasses some of these indeterminacies by unambiguously showing protein expression in the context of the three-dimensional ultrastructure of the cilium. Here, we apply immuno-SEM to specifically identify proteins on the primary cilia of mouse and human pancreatic islets, including post-translationally modified tubulin, intraflagellar transport (IFT)88, the small GTPase Arl13b, as well as subunits of axonemal dynein. Key parameters in sample preparation, immunolabeling and imaging acquisition are discussed to facilitate similar studies by others in the cilia research community.


Asunto(s)
Cilios , Islotes Pancreáticos , Cilios/ultraestructura , Cilios/metabolismo , Animales , Humanos , Ratones , Islotes Pancreáticos/ultraestructura , Islotes Pancreáticos/metabolismo , Microscopía Electrónica de Rastreo/métodos
2.
Lab Invest ; 102(1): 69-79, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34608240

RESUMEN

Mitochondrial homeostasis is crucial for the function of pancreatic ß-cells. ATP synthase inhibitory factor subunit 1 (IF1) is a mitochondrial protein interacting with ATP synthase to inhibit its enzyme activity. IF1 may also play a role in maintaining ATP synthase oligomerization and mitochondrial inner membrane formation. A recent study confirmed IF1 expresses in ß-cells. IF1 knockdown in cultured INS-1E ß-cells enhances glucose-induced insulin release. However, the role of IF1 in islet ß-cells remains little known. The present study investigates islets freshly isolated from mouse lines with global IF1 knockout (IF1-/-) and overexpression (OE). The glucose-stimulated insulin secretion was increased in islets from IF1-/- mice but decreased in islets from IF1 OE mice. Transmitted Electronic Microscopic assessment of isolated islets revealed that the number of matured insulin granules (with dense core) was relatively higher in IF1-/-, but fewer in IF1 OE islets than those of controlled islets. The mitochondrial ultrastructure within ß-cells of IF1 overexpressed islets was comparable with those of wild-type mice, whereas those in IF1-/- ß-cells showed increased mitochondrial mass. Mitochondrial network analysis in cultured INS-1 ß-cells showed a similar pattern with an increased mitochondrial network in IF1 knockdown cells. IF1 overexpressed INS-1 ß-cells showed a compromised rate of mitochondrial oxidative phosphorylation with attenuated cellular ATP content. In contrast, INS-1 cells with IF1 knockdown showed markedly increased cellular respiration with improved ATP production. These results support that IF1 is a negative regulator of insulin production and secretion via inhibiting mitochondrial mass and respiration in ß-cells. Therefore, inhibiting IF1 to improve ß-cell function in patients can be a novel therapeutic strategy to treat diabetes.


Asunto(s)
Homeostasis , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Proteínas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular Tumoral , Glucosa/farmacología , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Fosforilación Oxidativa , Proteínas/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteína Inhibidora ATPasa
3.
Peptides ; 147: 170704, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34826505

RESUMEN

The pancreatic islets contain beta-cells and alpha-cells, which are responsible for secreting two principal gluco-regulatory hormones; insulin and glucagon, respectively. However, they also contain delta-cells, a relatively sparse cell type that secretes somatostatin (SST). These cells have a complex morphology allowing them to establish an extensive communication network throughout the islet, despite their scarcity. Delta-cells are electrically excitable cells, and SST secretion is released in a glucose- and KATP-dependent manner. SST hyperpolarises the alpha-cell membrane and suppresses exocytosis. In this way, islet SST potently inhibits glucagon release. Recent studies investigating the activity of delta-cells have revealed they are electrically coupled to beta-cells via gap junctions, suggesting the delta-cell is more than just a paracrine inhibitor. In this Review, we summarize delta-cell morphology, function, and the role of SST signalling for regulating islet hormonal output. A distinguishing feature of this Review is that we attempt to use the discovery of this gap junction pathway, together with what is already known about delta-cells, to reframe the role of these cells in both health and disease. In particular, we argue that the discovery of gap junction communication between delta-cells and beta-cells provides new insights into the contribution of delta-cells to the islet hormonal defects observed in both type 1 and type 2 diabetes. This reappraisal of the delta-cell is important as it may offer novel insights into how the physiology of this cell can be utilised to restore islet function in diabetes.


Asunto(s)
Diabetes Mellitus/patología , Uniones Comunicantes/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Animales , Glucagón/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/ultraestructura , Somatostatina/metabolismo
4.
PLoS One ; 16(10): e0258208, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34614009

RESUMEN

Islet transplantation is being considered as an alternative treatment for type 1 diabetes. Despite recent progress, transplant recipients continue to experience progressive loss of insulin independence. Cyanidin-3-O-Glucoside (C3G) has shown to be protective against damage that may lead to post-transplant islet loss. In this study, human islets cultured with or without C3G were treated with human amylin, Aß1-42, H2O2, or rapamycin to mimic stresses encountered in the post-transplant environment. Samples of these islets were collected and assayed to determine C3G's effect on cell viability and function, reactive oxygen species (ROS), oxidative stress, amyloid formation, and the presence of inflammatory as well as autophagic markers. C3G treatment of human islets exposed to either amylin or Aß1-42 increased cell viability (p<0.01) and inhibited amyloid formation (p<0.01). A reduction in ROS and an increase in HO-1 gene expression as well as in vitro islet function were also observed in C3G-treated islets exposed to amylin or Aß1-42, although not significantly. Additionally, treatment with C3G resulted in a significant reduction in the protein expression of inflammatory markers IL-1ß and NLRP3 (p<0.01) as well as an increase in LC3 autophagic marker (p<0.05) in human islets treated with amylin, Aß1-42, rapamycin, or H2O2. Thus, C3G appears to have a multi-faceted protective effect on human islets in vitro, possibly through its anti-oxidant property and alteration of inflammatory as well as autophagic pathways.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Antocianinas/farmacología , Glucósidos/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/toxicidad , Islotes Pancreáticos/citología , Fragmentos de Péptidos/toxicidad , Adulto , Anciano , Autofagia/efectos de los fármacos , Biomarcadores/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Inflamación/patología , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/ultraestructura , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Adulto Joven
5.
PLoS One ; 16(8): e0255733, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34388180

RESUMEN

We previously reported that dye was effective to prevent the leakage of enzyme solutions from pancreatic glands during an islet isolation procedure. However, the dye used for islet isolation has not yet been optimized. In this study, we focused on pyoktanin blue (PB), diagnogreen (DG), and indigo carmine (IC) as potential candidates among clinically established dyes. A serial dilution assay was performed to determine minimal effective concentrations of each dye for detecting damaged pancreatic tissues. According to the outcome of serial dilution assays, double minimum effective concentrations of each dye were used for in vitro toxicity assays on islets and used in the isolation procedure to investigate whether they adversely affect islet isolation efficiency. The evaluations included islet yield, ADP/ATP, ATP/DNA, glucose stimulation test, and insulin/DNA assays. Islet viability cultured with PB contained medium was significantly lower than the other dyes. DG and IC appeared to be non-toxic to the islets. In isolation experiments, the islet yield in the DG group was considerably lower than that in the Control group, suggesting that DG might inhibit enzyme activity. The present study demonstrates that IC could be a promising candidate for an effective dye to detect damaged pancreatic tissues without affecting the enzyme activity and islet quality.


Asunto(s)
Colorantes/farmacología , Carmin de Índigo/farmacología , Islotes Pancreáticos/patología , Páncreas/efectos de los fármacos , Fenoles/farmacología , Pirroles/farmacología , Adenosina Difosfato/química , Adenosina Trifosfato/química , Animales , ADN/efectos de los fármacos , Glucosa/química , Humanos , Insulina/química , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/ultraestructura , Páncreas/patología , Páncreas/ultraestructura , Porcinos
6.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201511

RESUMEN

Diabetes is a metabolic disease that involves the death or dysfunction of the insulin-secreting ß cells in the pancreas. Consequently, most diabetes research is aimed at understanding the molecular and cellular bases of pancreatic development, islet formation, ß-cell survival, and insulin secretion. Complex interactions of signaling pathways and transcription factor networks regulate the specification, growth, and differentiation of cell types in the developing pancreas. Many of the same regulators continue to modulate gene expression and cell fate of the adult pancreas. The transcription factor NEUROD1 is essential for the maturation of ß cells and the expansion of the pancreatic islet cell mass. Mutations of the Neurod1 gene cause diabetes in humans and mice. However, the different aspects of the requirement of NEUROD1 for pancreas development are not fully understood. In this study, we investigated the role of NEUROD1 during the primary and secondary transitions of mouse pancreas development. We determined that the elimination of Neurod1 impairs the expression of key transcription factors for α- and ß-cell differentiation, ß-cell proliferation, insulin production, and islets of Langerhans formation. These findings demonstrate that the Neurod1 deletion altered the properties of α and ß endocrine cells, resulting in severe neonatal diabetes, and thus, NEUROD1 is required for proper activation of the transcriptional network and differentiation of functional α and ß cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Islotes Pancreáticos/citología , Páncreas/citología , Páncreas/embriología , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Diabetes Mellitus/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
Endocrinology ; 162(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34019639

RESUMEN

Tafazzin (TAZ) is a cardiolipin (CL) biosynthetic enzyme important for maintaining mitochondrial function. TAZ affects both the species and content of CL in the inner mitochondrial membrane, which are essential for normal cellular respiration. In pancreatic ß cells, mitochondrial function is closely associated with insulin secretion. However, the role of TAZ and CL in the secretion of insulin from pancreatic islets remains unknown. Male 4-month-old doxycycline-inducible TAZ knock-down (KD) mice and wild-type littermate controls were used. Immunohistochemistry was used to assess ß-cell morphology in whole pancreas sections, whereas ex vivo insulin secretion, CL content, RNA-sequencing analysis, and mitochondrial oxygen consumption were measured from isolated islet preparations. Ex vivo insulin secretion under nonstimulatory low-glucose concentrations was reduced ~52% from islets isolated from TAZ KD mice. Mitochondrial oxygen consumption under low-glucose conditions was also reduced ~58% in islets from TAZ KD animals. TAZ deficiency in pancreatic islets was associated with significant alteration in CL molecular species and elevated polyunsaturated fatty acid CL content. In addition, RNA-sequencing of isolated islets showed that TAZ KD increased expression of extracellular matrix genes, which are linked to pancreatic fibrosis, activated stellate cells, and impaired ß-cell function. These data indicate a novel role for TAZ in regulating pancreatic islet function, particularly under low-glucose conditions.


Asunto(s)
Aciltransferasas/deficiencia , Aciltransferasas/fisiología , Secreción de Insulina/fisiología , Islotes Pancreáticos/fisiología , Mitocondrias/fisiología , Aciltransferasas/genética , Animales , Cardiolipinas/análisis , Cardiolipinas/química , Doxiciclina/farmacología , Ácidos Grasos Insaturados/análisis , Femenino , Fibrosis , Técnicas de Silenciamiento del Gen , Islotes Pancreáticos/química , Islotes Pancreáticos/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxidación-Reducción , Consumo de Oxígeno/fisiología , Páncreas/patología
8.
Medicine (Baltimore) ; 100(17): e25642, 2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33907122

RESUMEN

ABSTRACT: Researchers divided the pancreas distal to the neck into 2 equal parts as the body and tail region by an arbitrary line. Surgeons considered the part of the pancreas, left to the aorta as the tail region. We performed this study to identify the transition zone of low-density to high-density islet cells for redefining the tail region.We quantified islets area proportion, beta-cell area proportion, and inter-islet distance in 9 Indian-adult-human non-diabetic pancreases from autopsy by using anti-synaptophysin and anti-insulin antibodies. Data were categorized under 3 regions like the proximal body, distal body, and distal part of the pancreas.Islet and beta-cell area proportion are progressively increased from head to tail region of the pancreas with a significant reduction in inter-islet distance and beta-cell percentage distal to the aorta. There is no significant difference in inter-islet distance and beta-cell percentage of the distal part of the body and tail region.Crowding of islets with intermingled microarchitecture begins in the pancreas distal to the aorta, which may be the beginning of the actual tail region. This study will provide insight into the preservation of islets-rich part of the pancreas during pancreatectomy and future prediction of new-onset diabetes.


Asunto(s)
Islotes Pancreáticos/ultraestructura , Páncreas/anatomía & histología , Páncreas/citología , Adulto , Autopsia , Metodologías Computacionales , Humanos , Inmunohistoquímica
9.
Sci Rep ; 10(1): 12187, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32699230

RESUMEN

Insulin gene mutations have been identified to cause monogenic diabetes, and most of which developed permanent neonatal diabetes at young ages before 6 months of age in humans. To establish an animal model of permanent diabetes, we performed genome editing using the CRISPR/Cas9 system. We generated a novel Kuma mutant mice with p.Q104del in the Insulin2 (Ins2) gene in a BRJ background that exhibits a severe immune deficiency. Kuma mutant mice are non-obese and developed hyperglycemia from 3 weeks after birth in both males and females, which are inherited in a dominant mode. Kuma mutant mice displayed reduced insulin protein levels from 3-weeks-old, which seem to be caused by the low stability of the mutant insulin protein. Kuma mutant showed a reduction in islet size and islet mass. Electron microscopic analysis revealed a marked decrease in the number and size of insulin granules in the beta-cells of the mutant mice. Hyperglycemia of the mutant can be rescued by insulin administration. Our results present a novel insulin mutation that causes permanent early-onset diabetes, which provides a model useful for islet transplantation studies.


Asunto(s)
Hiperglucemia/patología , Insulina/genética , Animales , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Femenino , Edición Génica , Prueba de Tolerancia a la Glucosa , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/genética , Insulina/uso terapéutico , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/fisiología , Islotes Pancreáticos/ultraestructura , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Electrónica , Oligodesoxirribonucleótidos/metabolismo , Tamaño de los Órganos
10.
Nat Commun ; 11(1): 2475, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424134

RESUMEN

Autoimmune ß-cell destruction leads to type 1 diabetes, but the pathophysiological mechanisms remain unclear. To help address this void, we created an open-access online repository, unprecedented in its size, composed of large-scale electron microscopy images ('nanotomy') of human pancreas tissue obtained from the Network for Pancreatic Organ donors with Diabetes (nPOD; www.nanotomy.org). Nanotomy allows analyses of complete donor islets with up to macromolecular resolution. Anomalies we found in type 1 diabetes included (i) an increase of 'intermediate cells' containing granules resembling those of exocrine zymogen and endocrine hormone secreting cells; and (ii) elevated presence of innate immune cells. These are our first results of mining the database and support recent findings that suggest that type 1 diabetes includes abnormalities in the exocrine pancreas that may induce endocrine cellular stress as a trigger for autoimmunity.


Asunto(s)
Bases de Datos como Asunto , Diabetes Mellitus Tipo 1/patología , Islotes Pancreáticos/ultraestructura , Microscopía Electrónica , Autoanticuerpos/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Granulocitos/inmunología , Humanos , Inmunidad Innata , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Donantes de Tejidos
11.
Biochem Biophys Res Commun ; 525(4): 1095-1102, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32184019

RESUMEN

Cell adhesion molecule L1-like protein (CHL1) is a member of neural recognition molecules of immunoglobulin superfamily primarily expressing in the nervous system. CHL1 regulates neuronal migration, axonal growth, and dendritic projection. Downregulation of CHL1 has been reported in ß cells of patients with type 2 diabetes (T2DM). However, the detailed role of CHL1 in ß cells has not been characterized. In this study, Real-Time PCR and Western blot were applied to investigate the tissue/cell distribution and expression of CHL1. Gain- or loss-of function studies were conducted in MIN6 cells to determine the effects of CHL1 on cell proliferation, apoptosis, cell cycle, and insulin secretion. Following silencing of CHL1 in MIN6 cells (si-CHL1), insulin secretion and the number of insulin secretary granules <50 nm from the cell membrane decreased in response to 20 mM glucose. Besides, silencing of CHL1 induced cell proliferation, reduced apoptosis, and prolonged S phase and shortened G1 phase of the cell cycle, contrary to overexpressing of CHL1. The inhibitor of ERK1/2MAPK eliminated the effect of CHL1 deficiency on the proliferation of MIN6 cells. In addition, high-fat diet could result in increased islet volume and ß cell proliferation, decreased CHL1 expression and activation of ERK pathway in mice islets. Consequently, CHL1 expression was decreased in islets of high-fat induced mice, which resulted in cell proliferation via ERK pathway and regulation of the cell cycle through p53 pathway. These mechanisms may contribute to pancreatic ß cell compensatory hyperplasia in obesity-induced pre-diabetes.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proliferación Celular/genética , Secreción de Insulina/genética , Islotes Pancreáticos/metabolismo , Animales , Apoptosis/genética , Moléculas de Adhesión Celular/genética , Ciclo Celular/genética , Dieta Alta en Grasa , Silenciador del Gen , Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/ultraestructura , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Regulación hacia Arriba
12.
J Diabetes Complications ; 34(3): 107497, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31866258

RESUMEN

AIMS: There is a pancreatic islet adaptation in obese subjects, resulting in insulin resistance and diabetes type 2. We studied the effect of intermittent fasting (IntF) on the islet structure of diet-induced obese (DIO) mice. METHODS: Three-month-old male mice fed a control diet (C, 10% Kcal fat) or a high-fat diet (HF, 50% Kcal fat) for two months (n = 20 each group). Then, half of each group did IntF (alternating 24 h fed/24 h fast), continuing in their diets four more weeks: C, C-IntF, HF, HF-IntF. Islets were prepared to microscopy or isolated for molecular analysis. RESULTS: HF group (vs. C group) showed hyperglycemia, hyperinsulinemia, hyperleptinemia, hypoadiponectinemia, glucose intolerance, insulin resistance, and islet hypertrophy with a consequent higher both the alpha-cell and beta-cell masses. In the HF group (vs. C), there was low PDX1 (pancreatic and duodenal homeobox 1), and IntF did not alter PDX1. There was a low p-AKT/AKT ratio (protein kinase B), and IntF enhanced it. Also, tumor suppressor p53 was increased, and IntF decreased it. IL (interleukin) -6 was higher in the HF group (vs. C), and HF-IntF (vs. C-IntF). Any significant change in NFkB was seen among groups. CONCLUSIONS: IntF improves pancreatic islet structure in DIO mice, even with continued HF diet intake, primarily considering on the alpha- and beta-cell masses regulation, then improving insulin signaling and decreasing cell apoptosis. Future research should explore whether the shortening of the IntF extend could maintain the benefits observed in the long term.


Asunto(s)
Ayuno/fisiología , Células Secretoras de Glucagón/patología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Obesidad/patología , Animales , Recuento de Células , Proliferación Celular , Microambiente Celular/fisiología , Dieta Alta en Grasa , Islotes Pancreáticos/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/etiología
13.
Diabetes ; 69(3): 342-354, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31836690

RESUMEN

Human but not mouse islets transplanted into immunodeficient NSG mice effectively accumulate lipid droplets (LDs). Because chronic lipid exposure is associated with islet ß-cell dysfunction, we investigated LD accumulation in the intact human and mouse pancreas over a range of ages and states of diabetes. Very few LDs were found in normal human juvenile pancreatic acinar and islet cells, with numbers subsequently increasing throughout adulthood. While accumulation appeared evenly distributed in postjuvenile acinar and islet cells in donors without diabetes, LDs were enriched in islet α- and ß-cells from donors with type 2 diabetes (T2D). LDs were also found in the islet ß-like cells produced from human embryonic cell-derived ß-cell clusters. In contrast, LD accumulation was nearly undetectable in the adult rodent pancreas, even in hyperglycemic and hyperlipidemic models or 1.5-year-old mice. Taken together, there appear to be significant differences in pancreas islet cell lipid handling between species, and the human juvenile and adult cell populations. Moreover, our results suggest that LD enrichment could be impactful to T2D islet cell function.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Células Secretoras de Glucagón/patología , Células Secretoras de Insulina/patología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/patología , Gotas Lipídicas/patología , Células Acinares/patología , Células Acinares/ultraestructura , Adolescente , Adulto , Factores de Edad , Anciano , Animales , Niño , Preescolar , Diabetes Mellitus Experimental/patología , Células Madre Embrionarias , Femenino , Células Secretoras de Glucagón/ultraestructura , Humanos , Lactante , Células Secretoras de Insulina/ultraestructura , Islotes Pancreáticos/citología , Islotes Pancreáticos/ultraestructura , Gotas Lipídicas/ultraestructura , Masculino , Ratones , Microscopía Electrónica , Microscopía Fluorescente , Persona de Mediana Edad , Ratas , Donantes de Tejidos , Adulto Joven
14.
Nat Commun ; 10(1): 3700, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31420552

RESUMEN

Little is known about the role of islet delta cells in regulating blood glucose homeostasis in vivo. Delta cells are important paracrine regulators of beta cell and alpha cell secretory activity, however the structural basis underlying this regulation has yet to be determined. Most delta cells are elongated and have a well-defined cell soma and a filopodia-like structure. Using in vivo optogenetics and high-speed Ca2+ imaging, we show that these filopodia are dynamic structures that contain a secretory machinery, enabling the delta cell to reach a large number of beta cells within the islet. This provides for efficient regulation of beta cell activity and is modulated by endogenous IGF-1/VEGF-A signaling. In pre-diabetes, delta cells undergo morphological changes that may be a compensation to maintain paracrine regulation of the beta cell. Our data provides an integrated picture of how delta cells can modulate beta cell activity under physiological conditions.


Asunto(s)
Islotes Pancreáticos/ultraestructura , Comunicación Paracrina , Estado Prediabético/patología , Seudópodos/ultraestructura , Células Secretoras de Somatostatina/ultraestructura , Animales , Glucemia/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Microscopía Intravital , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ratones , Ratones Transgénicos , Microscopía Electrónica , Imagen Óptica , Optogenética , Estado Prediabético/metabolismo , Seudópodos/metabolismo , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Sci Rep ; 9(1): 8449, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186447

RESUMEN

The pancreatic islet is a complex micro-organ containing numerous cell types, including endocrine, immune, and endothelial cells. The communication of these systems is lost upon isolation of the islets, and therefore the pathogenesis of diabetes can only be fully understood by studying this organized, multicellular environment in vivo. We have developed several adaptable tools to create a versatile platform to interrogate ß-cell function in vivo. Specifically, we developed ß-cell-selective virally-encoded fluorescent protein biosensors that can be rapidly and easily introduced into any mouse. We then coupled the use of these biosensors with intravital microscopy, a powerful tool that can be used to collect cellular and subcellular data from living tissues. Together, these approaches allowed the observation of in vivo ß-cell-specific ROS dynamics using the Grx1-roGFP2 biosensor and calcium signaling using the GcAMP6s biosensor. Next, we utilized abdominal imaging windows (AIW) to extend our in vivo observations beyond single-point terminal measurements to collect longitudinal physiological and biosensor data through repeated imaging of the same mice over time. This platform represents a significant advancement in our ability to study ß-cell structure and signaling in vivo, and its portability for use in virtually any mouse model will enable meaningful studies of ß-cell physiology in the endogenous islet niche.


Asunto(s)
Células Endoteliales/ultraestructura , Células Secretoras de Insulina/ultraestructura , Microscopía Intravital/métodos , Islotes Pancreáticos/ultraestructura , Animales , Técnicas Biosensibles , Señalización del Calcio/genética , Señalización del Calcio/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos , Ratones
16.
Cell Biol Int ; 43(8): 846-851, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31115951

RESUMEN

Lelio Orci has made seminal contributions to our understanding of pancreatic islet structure and function. He introduced quantitative criteria to structural analysis in the study of endocrine pancreas in a series of works performed in collaboration with Albert Renold, Roger Unger, and Donald Steiner. Orci has moved islet cell morphology from the primitive era of histochemistry and electron microscopy into the modern era of cell biology, applying the most advanced techniques and covering every aspect of normal and pathological structure-function relationships. In collaboration with James Rothman in New York and Randy Schekman in Berkley, Orci discovered that the transport steps from the endoplasmic reticulum to the Golgi complex, and within the Golgi, are mediated by two sets of vesicles coated with protein envelopes different from clathrin.


Asunto(s)
Retículo Endoplásmico , Aparato de Golgi , Islotes Pancreáticos , Distinciones y Premios , Transporte Biológico , Clatrina/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Historia del Siglo XX , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/ultraestructura , Italia , Suiza
17.
Cell Metab ; 29(3): 769-783.e4, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30713110

RESUMEN

The interaction between the immune system and endocrine cells in the pancreas is crucial for the initiation and progression of type 1 diabetes (T1D). Imaging mass cytometry (IMC) enables multiplexed assessment of the abundance and localization of more than 30 proteins on the same tissue section at 1-µm resolution. Herein, we have developed a panel of 33 antibodies that allows for the quantification of key cell types including pancreatic exocrine cells, islet cells, immune cells, and stromal components. We employed this panel to analyze 12 pancreata obtained from donors with clinically diagnosed T1D and 6 pancreata from non-diabetic controls. In the pancreata from donors with T1D, we simultaneously visualized significant alterations in islet architecture, endocrine cell composition, and immune cell presentation. Indeed, we demonstrate the utility of IMC to investigate complex events on the cellular level that will provide new insights on the pathophysiology of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Citometría de Imagen/métodos , Islotes Pancreáticos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Células Endocrinas/inmunología , Células Endocrinas/metabolismo , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura
18.
J Histochem Cytochem ; 67(1): 41-52, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29771178

RESUMEN

Islet microvasculature provides key architectural and functional roles, yet the morphological features of islets from patients with type 1 diabetes are poorly defined. We examined islet and exocrine microvasculature networks by multiplex immunofluorescence imaging of pancreases from organ donors with and without type 1 diabetes (n=17 and n=16, respectively) and determined vessel diameter, density, and area. We also analyzed these variables in insulin-positive and insulin-negative islets of 7 type 1 diabetes donors. Control islet vessel diameter was significantly larger (7.6 ± 1.1 µm) compared with vessels in diabetic islets (6.2 ± 0.8 µm; p<0.001). Control islet vessel density (number/islet) was significantly lower (5.3 ± 0.6) versus diabetic islets (9.3 ± 0.2; p<0.001). Exocrine vessel variables were not significantly different between groups. Islets with residual beta-cells were comparable to control islets for both vessel diameter and density and were significantly different from insulin-negative islets within diabetic donors (p<0.05). Islet smooth muscle actin area had a significant positive correlation with age in both groups (p<0.05), which could negatively impact islet transplantation efficiency from older donors. These data underscore the critical relationship of islet beta-cells and islet vessel morphology in type 1 diabetes. These studies provide new knowledge of the islet microvasculature in diabetes and aging.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/patología , Microvasos/patología , Actinas/análisis , Adolescente , Adulto , Niño , Femenino , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Insulina/análisis , Células Secretoras de Insulina/ultraestructura , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/patología , Islotes Pancreáticos/ultraestructura , Masculino , Microscopía Confocal/métodos , Microvasos/ultraestructura , Donantes de Tejidos , Adulto Joven
19.
Toxicol Pathol ; 47(1): 35-43, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30407147

RESUMEN

We characterized pancreatic islet lesions induced by several quinolones using functional and morphological examinations of the pancreatic islets in male rats orally administered gatifloxacin, lomefloxacin, or levofloxacin at 300 mg/kg for 14 consecutive days. Consequently, in contrast to lomefloxacin or levofloxacin, gatifloxacin increased serum glucose and glycosylated albumin on day 14 and elevated serum glucose tended to decrease insulin in the intravenous glucose tolerance test. Microscopically, only gatifloxacin induced cytoplasmic vacuoles containing eosinophilic homogenous contents in islet cells. Immunohistochemical examination revealed that vacuolated islet cells were positively stained for insulin, demonstrating they were pancreatic ß cells. Electron microscopy showed that the cytoplasmic vacuoles represented dilated cisterna of the rough endoplasmic reticulum filled with electron-lucent materials in pancreatic ß cells. Moreover, insulin secretory granules were drastically decreased in vacuolated islet cells, suggesting impaired insulin synthesis and/or transport. This gatifloxacin-induced pancreatic toxicity in rats was considered to be associated with high pancreatic drug distribution. These results demonstrated that gatifloxacin provoked functional and morphological pancreatic ß cell alteration associated with impaired insulin synthesis and/or transport, leading to hyperglycemia.


Asunto(s)
Antibacterianos/toxicidad , Gatifloxacina/toxicidad , Islotes Pancreáticos/efectos de los fármacos , Administración Oral , Animales , Antibacterianos/sangre , Citoplasma/efectos de los fármacos , Citoplasma/ultraestructura , Gatifloxacina/sangre , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/biosíntesis , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Masculino , Ratas Sprague-Dawley , Distribución Tisular
20.
Methods Mol Biol ; 1916: 223-232, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30535699

RESUMEN

The islets of Langerhans release vital hormones involved in the regulation of blood sugar and other aspects of metabolism. The islets are housed in diffuse clusters of cells within the exocrine pancreas and, therefore, purification of these cells for research or transplant purposes is a difficult undertaking. Here, a detailed protocol is presented for purification of islets from rat pancreas using limited collagenase digestion and step gradient centrifugation techniques. In addition, a method for assessing islet viability is presented using perifusion under both basal and stimulatory glucose conditions, with measurement of the hormone released using an immunoassay for insulin.


Asunto(s)
Separación Celular/métodos , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/ultraestructura , Páncreas/metabolismo , Animales , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Páncreas/patología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA