Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 939
Filtrar
1.
J Biol Chem ; 299(7): 104836, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37209827

RESUMEN

Insulin is made from proinsulin, but the extent to which fasting/feeding controls the homeostatically regulated proinsulin pool in pancreatic ß-cells remains largely unknown. Here, we first examined ß-cell lines (INS1E and Min6, which proliferate slowly and are routinely fed fresh medium every 2-3 days) and found that the proinsulin pool size responds to each feeding within 1 to 2 h, affected both by the quantity of fresh nutrients and the frequency with which they are provided. We observed no effect of nutrient feeding on the overall rate of proinsulin turnover as quantified from cycloheximide-chase experiments. We show that nutrient feeding is primarily linked to rapid dephosphorylation of translation initiation factor eIF2α, presaging increased proinsulin levels (and thereafter, insulin levels), followed by its rephosphorylation during the ensuing hours that correspond to a fall in proinsulin levels. The decline of proinsulin levels is blunted by the integrated stress response inhibitor, ISRIB, or by inhibition of eIF2α rephosphorylation with a general control nonderepressible 2 (not PERK) kinase inhibitor. In addition, we demonstrate that amino acids contribute importantly to the proinsulin pool; mass spectrometry shows that ß-cells avidly consume extracellular glutamine, serine, and cysteine. Finally, we show that in both rodent and human pancreatic islets, fresh nutrient availability dynamically increases preproinsulin, which can be quantified without pulse-labeling. Thus, the proinsulin available for insulin biosynthesis is rhythmically controlled by fasting/feeding cycles.


Asunto(s)
Células Secretoras de Insulina , Nutrientes , Proinsulina , Humanos , Insulina/biosíntesis , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Nutrientes/farmacología , Proinsulina/biosíntesis , Proinsulina/metabolismo , Estrés Fisiológico , Transducción de Señal , Línea Celular , Regulación hacia Arriba
2.
Crit Rev Eukaryot Gene Expr ; 32(6): 33-46, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35997116

RESUMEN

Studies have reported that miRNAs regulate ß-cell differentiation, pancreatic development, and insulin secretion. However, the biological function of miRNAs during the formation of insulin-producing cells (IPCs) from umbilical cord-derived mesenchymal stem cells (UCMSCs) is poorly understood. Herein, the role and mechanism of miR-200b-3p during UCMSC differentiation into IPCs were investigated. UCMSCs were induced for IPC differentiation. An animal model was established by transplanting UCMSC-derived IPCs into streptozotocin-induced diabetic mice. Cell surface markers of undifferentiated UCMSCs and the expression of proinsulin and Pdx-1 in UCMSC-derived IPCs were measured by flow cytometry analysis. The interaction between miR-200b-3p and zinc finger E-box binding homeobox 2 (ZEB2) 3' untranslated region (UTR) was confirmed by luciferase reporter assay. Insulin secretion in UCMSC-derived IPCs was measured by enzyme-linked immunosorbent assay (ELISA). Islet marker (insulin and Pdx-1) levels were evaluated using immunofluorescence staining. In this study, undifferentiated UCMSCs had MSC phenotype and the potential for osteogenesis and adipogenesis. UCMSC-derived IPCs displayed glucose responsive insulin secretion and expressed insulin, Pdx-1 and proinsulin. miR-200b-3p was overexpressed in UCMSC-derived IPCs. Mechanically, miR-200b-3p targeted ZEB2. ZEB2 knockdown reversed the inhibitory effect of miR-200b-3p downregulation on IPC differentiation from UCMSCs in vitro. Moreover, miR-200b-3p silencing inhibited the anti-hypoglycemic effects and insulinogenesis of UCMSC-derived IPCs grafts in the kidney capsule of diabetic mice. Overall, miR-200b-3p induces the formation of IPCs from UCMSCs by targeting ZEB2.


Asunto(s)
Diabetes Mellitus Experimental , Células Madre Mesenquimatosas , MicroARNs , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Animales , Diabetes Mellitus Experimental/genética , Insulina/biosíntesis , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proinsulina , Cordón Umbilical/citología , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética
3.
Science ; 376(6599): 1321-1327, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35709255

RESUMEN

The emergence of new therapeutic modalities requires complementary tools for their efficient syntheses. Availability of methodologies for site-selective modification of biomolecules remains a long-standing challenge, given the inherent complexity and the presence of repeating residues that bear functional groups with similar reactivity profiles. We describe a bioconjugation strategy for modification of native peptides relying on high site selectivity conveyed by enzymes. We engineered penicillin G acylases to distinguish among free amino moieties of insulin (two at amino termini and an internal lysine) and manipulate cleavable phenylacetamide groups in a programmable manner to form protected insulin derivatives. This enables selective and specific chemical ligation to synthesize homogeneous bioconjugates, improving yield and purity compared to the existing methods, and generally opens avenues in the functionalization of native proteins to access biological probes or drugs.


Asunto(s)
Insulina , Penicilina Amidasa , Péptidos , Ingeniería de Proteínas , Secuencia de Aminoácidos , Humanos , Insulina/análogos & derivados , Insulina/biosíntesis , Lisina/química , Penicilina Amidasa/química , Penicilina Amidasa/genética , Péptidos/química , Péptidos/genética , Ingeniería de Proteínas/métodos
4.
Connect Tissue Res ; 63(5): 498-513, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35129018

RESUMEN

PURPOSE: Type V collagen (collagen V) is one of the important components of extracellular matrix (ECM) in pancreas. We previously reported that pre-coating collagen V on the culture dishes enhanced insulin production in INS-1 rat pancreatic ß cells. In this study, we investigate the underlying mechanism. RESULTS: Insulin biosynthesis and secretion are both increased in INS-1 cells cultured on collagen V-coated dishes, accompanied by the reduced nuclear translocation of Yes-associated protein (YAP), a transcriptional co-activator. YAP, the downstream effector of Hippo signaling pathway, plays an important role in the development and function of pancreas. Inhibition of YAP activation by verteporfin further up-regulates insulin biosynthesis and secretion. Silencing large tumor suppressor (LATS), a core component of Hippo pathway which inhibits activity of YAP by phosphorylation, by siRNA transfection inhibits both insulin biosynthesis and secretion. In the present study, the protein level of insulin-like growth factor 1 receptor (IGF-1 R), detected as the upstream molecule of YAP, is reduced in the INS-1 cells cultured on the dishes coated with collagen V. The silencing of IGF-1 R by siRNA transfection further enhances insulin biosynthesis and secretion. IGF-1 treatment reduces collagen V-induced up-regulation of insulin biosynthesis and secretion, accompanying the increased nuclear YAP. CONCLUSION: Inhibition of IGF-1 R/YAP signal pathway is involved in collagen V-induced insulin biosynthesis and secretion in INS-1 cells.


Asunto(s)
Insulina , Islotes Pancreáticos , Receptor IGF Tipo 1 , Transducción de Señal , Proteínas Señalizadoras YAP , Animales , Colágeno Tipo V/farmacología , Insulina/biosíntesis , Islotes Pancreáticos/metabolismo , Fosforilación , ARN Interferente Pequeño/metabolismo , Ratas , Receptor IGF Tipo 1/metabolismo , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP/metabolismo
5.
Cells ; 10(12)2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34944065

RESUMEN

Despite the immense functional relevance of GPR56 (gene ADGRG1) in highly diverse (patho)physiological processes such as tumorigenesis, immune regulation, and brain development, little is known about its exact tissue localization. Here, we validated antibodies for GPR56-specific binding using cells with tagged GPR56 or eliminated ADGRG1 in immunotechniques. Using the most suitable antibody, we then established the human GPR56 tissue expression profile. Overall, ADGRG1 RNA-sequencing data of human tissues and GPR56 protein expression correlate very well. In the adult brain especially, microglia are GPR56-positive. Outside the central nervous system, GPR56 is frequently expressed in cuboidal or highly prismatic secreting epithelia. High ADGRG1 mRNA, present in the thyroid, kidney, and placenta is related to elevated GPR56 in thyrocytes, kidney tubules, and the syncytiotrophoblast, respectively. GPR56 often appears in association with secreted proteins such as pepsinogen A in gastric chief cells and insulin in islet ß-cells. In summary, GPR56 shows a broad, not cell-type restricted expression in humans.


Asunto(s)
Carcinogénesis/genética , Insulina/genética , Neoplasias/genética , Receptores Acoplados a Proteínas G/genética , Adhesión Celular/genética , Células Principales Gástricas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Insulina/biosíntesis , Islotes Pancreáticos/metabolismo , Riñón/metabolismo , Microglía/metabolismo , Microglía/patología , Neoplasias/patología , Pepsinógeno A/biosíntesis , Pepsinógeno A/genética , Placenta/metabolismo , Embarazo , RNA-Seq , Glándula Tiroides/metabolismo
6.
Molecules ; 26(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34770916

RESUMEN

The aim of our study was to investigate the effect of three lignans (schisandrol A, schisandrol B, and schisandrin C) on insulin secretion in rat INS-1 pancreatic ß-cells and glucose uptake in mouse C2C12 skeletal muscle cells. Schisandrol A and schisandrin C enhanced insulin secretion in response to high glucose levels with no toxic effects on INS-1 cells. The effect of schisandrin C was superior to that of gliclazide (positive control), a drug commonly used to treat type 2 diabetes (T2D). In addition, western blot analysis showed that the expression of associated proteins, including peroxisome proliferator-activated receptor γ (PPARγ), pancreatic and duodenal homeobox 1 (PDX-1), phosphatidylinositol 3-kinase (PI3K), Akt, and insulin receptor substrate-2 (IRS-2), was increased in INS-1 cells after treatment with schisandrin C. In addition, insulin secretion effect of schisandrin C were enhanced by the Bay K 8644 (L-type Ca2+ channel agonist) and glibenclamide (K+ channel blocker), were abolished by the nifedipine (L-type Ca2+ channel blocker) and diazoxide (K+ channel activator). Moreover, schisandrin C enhanced glucose uptake with no toxic effects on C2C12 cells. Western blot analysis showed that the expression of associated proteins, including insulin receptor substrate-1 (IRS-1), AMP-activated protein kinase (AMPK), PI3K, Akt, glucose transporter type 4 (GLUT-4), was increased in C2C12 cells after treatment with schisandrin C. Schisandrin C may improve hyperglycemia by enhancing insulin secretion in pancreatic ß-cells and improving glucose uptake into skeletal muscle cells. Our findings may provide evidence that schisandrin C may be beneficial in devising novel anti-T2D strategies.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , Lignanos/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Compuestos Policíclicos/farmacología , Adenosina Trifosfato/biosíntesis , Biomarcadores , Canales de Calcio/genética , Canales de Calcio/metabolismo , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Línea Celular , Ciclooctanos/química , Ciclooctanos/farmacología , Expresión Génica , Lignanos/química , Compuestos Policíclicos/química , Canales de Potasio/genética , Canales de Potasio/metabolismo
7.
Curr Drug Metab ; 22(13): 1017-1034, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34825869

RESUMEN

The N-3 polyunsaturated fatty acids (PUFAs) have a wide range of health benefits, including antiinflammatory effects, improvements in lipids metabolism and promoting insulin secretion, as well as reduction of cancer risk. Numerous studies support that N-3 PUFAs have the potentials to improve many metabolic diseases, such as diabetes, nonalcoholic fatty liver disease and obesity, which are attributable to N-3 PUFAs mediated enhancement of insulin secretion by pancreatic ß-cells and improvements in insulin sensitivity and metabolic disorders in peripheral insulin-sensitive tissues such as liver, muscles, and adipose tissue. In this review, we summarized the up-to-date clinical and basic studies on the regulatory effects and molecular mechanisms of N-3 PUFAs mediated benefits on pancreatic ß-cells, adipose tissue, liver, and muscles in the context of glucose and/or lipid metabolic disorders. We also discussed the potential factors involved in the inconsistent results from different clinical researches of N-3 PUFAs.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Células Secretoras de Insulina , Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Enfermedades Metabólicas , Animales , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-3/farmacología , Humanos , Insulina/biosíntesis , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Enfermedades Metabólicas/clasificación , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/prevención & control
8.
Front Endocrinol (Lausanne) ; 12: 754693, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659132

RESUMEN

Insight into folding mechanisms of proinsulin has been provided by analysis of dominant diabetes-associated mutations in the human insulin gene (INS). Such mutations cause pancreatic ß-cell dysfunction due to toxic misfolding of a mutant proinsulin and impairment in trans of wild-type insulin secretion. Anticipated by the "Akita" mouse (a classical model of monogenic diabetes mellitus; DM), this syndrome illustrates the paradigm endoreticulum (ER) stress leading to intracellular proteotoxicity. Diverse clinical mutations directly or indirectly perturb native disulfide pairing leading to protein misfolding and aberrant aggregation. Although most introduce or remove a cysteine (Cys; leading in either case to an unpaired thiol group), non-Cys-related mutations identify key determinants of folding efficiency. Studies of such mutations suggest that the hormone's evolution has been constrained not only by structure-function relationships, but also by the susceptibility of its single-chain precursor to impaired foldability. An intriguing hypothesis posits that INS overexpression in response to peripheral insulin resistance likewise leads to chronic ER stress and ß-cell dysfunction in the natural history of non-syndromic Type 2 DM. Cryptic contributions of conserved residues to folding efficiency, as uncovered by rare genetic variants, define molecular links between biophysical principles and the emerging paradigm of Darwinian medicine: Biosynthesis of proinsulin at the edge of non-foldability provides a key determinant of "diabesity" as a pandemic disease of civilization.


Asunto(s)
Diabetes Mellitus/genética , Proinsulina/genética , Pliegue de Proteína , Evolución Molecular , Humanos , Insulina/biosíntesis , Mutación
9.
Nat Commun ; 12(1): 4458, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294685

RESUMEN

The cellular identity of pancreatic polypeptide (Ppy)-expressing γ-cells, one of the rarest pancreatic islet cell-type, remains elusive. Within islets, glucagon and somatostatin, released respectively from α- and δ-cells, modulate the secretion of insulin by ß-cells. Dysregulation of insulin production raises blood glucose levels, leading to diabetes onset. Here, we present the genetic signature of human and mouse γ-cells. Using different approaches, we identified a set of genes and pathways defining their functional identity. We found that the γ-cell population is heterogeneous, with subsets of cells producing another hormone in addition to Ppy. These bihormonal cells share identity markers typical of the other islet cell-types. In mice, Ppy gene inactivation or conditional γ-cell ablation did not alter glycemia nor body weight. Interestingly, upon ß-cell injury induction, γ-cells exhibited gene expression changes and some of them engaged insulin production, like α- and δ-cells. In conclusion, we provide a comprehensive characterization of γ-cells and highlight their plasticity and therapeutic potential.


Asunto(s)
Insulina/biosíntesis , Células Secretoras de Polipéptido Pancreático/metabolismo , Polipéptido Pancreático/metabolismo , Precursores de Proteínas/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal , Linaje de la Célula/genética , Femenino , Técnicas de Sustitución del Gen , Humanos , Células Secretoras de Insulina/clasificación , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Páncreas/citología , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Polipéptido Pancreático/deficiencia , Polipéptido Pancreático/genética , Células Secretoras de Polipéptido Pancreático/clasificación , Células Secretoras de Polipéptido Pancreático/citología , Embarazo , RNA-Seq
10.
Sci Rep ; 11(1): 12409, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117315

RESUMEN

The trend of regenerative therapy for diabetes in human and veterinary practices has conceptually been proven according to the Edmonton protocol and animal models. Establishing an alternative insulin-producing cell (IPC) resource for further clinical application is a challenging task. This study investigated IPC generation from two practical canine mesenchymal stem cells (cMSCs), canine bone marrow-derived MSCs (cBM-MSCs) and canine adipose-derived MSCs (cAD-MSCs). The results illustrated that cBM-MSCs and cAD-MSCs contain distinct pancreatic differentiation potential and require the tailor-made induction protocols. The effective generation of cBM-MSC-derived IPCs needs the integration of genetic and microenvironment manipulation using a hanging-drop culture of PDX1-transfected cBM-MSCs under a three-step pancreatic induction protocol. However, this protocol is resource- and time-consuming. Another study on cAD-MSC-derived IPC generation found that IPC colonies could be obtained by a low attachment culture under the three-step induction protocol. Further, Notch signaling inhibition during pancreatic endoderm/progenitor induction yielded IPC colonies through the trend of glucose-responsive C-peptide secretion. Thus, this study showed that IPCs could be obtained from cBM-MSCs and cAD-MSCs through different induction techniques. Also, further signaling manipulation studies should be conducted to maximize the protocol's efficiency.


Asunto(s)
Tejido Adiposo/metabolismo , Células de la Médula Ósea/metabolismo , Insulina/biosíntesis , Células Madre Mesenquimatosas/citología , Animales , Adhesión Celular , Técnicas de Cultivo de Célula , Células Cultivadas , Perros , Células Madre Mesenquimatosas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
11.
FASEB J ; 35(5): e21515, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811688

RESUMEN

The conserved endoplasmic reticulum (ER) membrane protein TRAPα (translocon-associated protein, also known as signal sequence receptor 1, SSR1) has been reported to play a critical but unclear role in insulin biosynthesis. TRAPα/SSR1 is one component of a four-protein complex including TRAPß/SSR2, TRAPγ/SSR3, and TRAPδ/SSR4. The TRAP complex topologically has a small exposure on the cytosolic side of the ER via its TRAPγ/SSR3 subunit, whereas TRAPß/SSR2 and TRAPδ/SSR4 function along with TRAPα/SSR1 largely on the luminal side of the ER membrane. Here, we have examined pancreatic ß-cells with deficient expression of either TRAPß/SSR2 or TRAPδ/SSR4, which does not perturb mRNA expression levels of other TRAP subunits, or insulin mRNA. However, deficient protein expression of TRAPß/SSR2 and, to a lesser degree, TRAPδ/SSR4, diminishes the protein levels of other TRAP subunits, concomitant with deficient steady-state levels of proinsulin and insulin. Deficient TRAPß/SSR2 or TRAPδ/SSR4 is not associated with any apparent defect of exocytotic mechanism but rather by a decreased abundance of the proinsulin and insulin that accompanies glucose-stimulated secretion. Amino acid pulse labeling directly establishes that much of the steady-state deficiency of intracellular proinsulin can be accounted for by diminished proinsulin biosynthesis, observed in a pulse-labeling as short as 5 minutes. The proinsulin and insulin levels in TRAPß/SSR2 or TRAPδ/SSR4 null mutant ß-cells are notably recovered upon re-expression of the missing TRAP subunit, accompanying a rebound of proinsulin biosynthesis. Remarkably, overexpression of TRAPα/SSR1 can also suppress defects in ß-cells with diminished expression of TRAPß/SSR2, strongly suggesting that TRAPß/SSR2 is needed to support TRAPα/SSR1 function.


Asunto(s)
Proteínas de Unión al Calcio/deficiencia , Retículo Endoplásmico/metabolismo , Glucosa/metabolismo , Insulina/biosíntesis , Insulinoma/patología , Glicoproteínas de Membrana/deficiencia , Proinsulina/biosíntesis , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores de Péptidos/deficiencia , Animales , Células Cultivadas , Células Secretoras de Insulina/citología , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Ratas
12.
Mol Cell Endocrinol ; 526: 111193, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33610643

RESUMEN

Evidence is presented for expression of the insulin receptor on the surface of mammalian spermatozoa as well as transcripts for the receptor substrate adaptor proteins (IRS1-4) needed to mediate insulin action. Exposure to this hormone resulted in insulin receptor phosphorylation (pTyr972), activation of AKT (pSer473) and the stimulation of sperm motility. Intriguingly, the male germ line is also shown to be capable of generating insulin, possessing the relevant mRNA transcript and expressing strong immunocytochemical signals for both insulin and C-peptide. Insulin could be released from the spermatozoa by sonication in a concentration-dependent manner but was not secreted in response to glucose, fructose or stimulation with progesterone. However, insulin release could be induced by factors present in human uterine lavages. Furthermore, the endometrium was also shown to possess the machinery for insulin production and action (mRNA, insulin, C-peptide, proprotein convertase and insulin receptor), releasing insulin into the uterine lumen prior to ovulation. These studies emphasize the fundamental importance of extra-pancreatic insulin in regulating the reproductive process, particularly in the support of spermatozoa on their perilous voyage to the site of fertilization.


Asunto(s)
Insulina/biosíntesis , Páncreas/metabolismo , Espermatozoides/citología , Espermatozoides/metabolismo , Animales , Péptido C/metabolismo , Supervivencia Celular , Endometrio/metabolismo , Epitelio/metabolismo , Femenino , Células Germinativas/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Secreción de Insulina , Masculino , Mamíferos/metabolismo , Ratones , Isoformas de Proteínas/metabolismo , Ratas , Receptor de Insulina/metabolismo , Útero/metabolismo
13.
Peptides ; 135: 170434, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33172827

RESUMEN

To study whether activation of GLP-1 receptors importantly contributes to the insulinotropic action of exogenously administered glucagon, we have performed whole animal experiments in normal mice and in mice with GLP-1 receptor knockout. Glucagon (1, 3 or 10 µg/kg), the GLP-1 receptor antagonist exendin 9-39 (30 nmol/kg), glucose (0.35 g/kg) or the incretin hormone glucose-dependent insulinotropic polypeptide (GIP; 3 nmol/kg) was injected intravenously or glucose (75 mg) was given orally through gavage. Furthermore, islets were isolated and incubated in the presence of glucose with or without glucagon. It was found that the insulin response to intravenous glucagon was preserved in GLP-1 receptor knockout mice but that glucagon-induced insulin secretion was markedly suppressed in islets from GLP-1 receptor knockout mice. Similarly, the GLP-1 receptor antagonist markedly suppressed glucagon-induced insulin secretion in wildtype mice. These data suggest that GLP-1 receptors contribute to the insulinotropic action of glucagon and that there is a compensatory mechanism in GLP-1 receptor knockout mice that counteracts a reduced effect of glucagon. Two potential compensatory mechanisms (glucose and GIP) were explored. However, neither of these seemed to explain why the insulin response to glucagon is not suppressed in GLP-1 receptor knockout mice. Based on these data we confirm the hypothesis that glucagon-induced insulin secretion is partially mediated by GLP-1 receptors on the beta cells and we propose that a compensatory mechanism, the nature of which remains to be established, is induced in GLP-1 receptor knockout mice to counteract the expected impaired insulin response to glucagon in these mice.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/genética , N-Metiltransferasa de Histona-Lisina/genética , Secreción de Insulina/genética , Insulina/biosíntesis , Animales , Glucemia , Modelos Animales de Enfermedad , Glucagón/antagonistas & inhibidores , Glucagón/farmacología , Glucosa/farmacología , Humanos , Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Fragmentos de Péptidos/farmacología
14.
BMJ Case Rep ; 13(11)2020 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-33168531

RESUMEN

A 27-year-old otherwise healthy man of African descent presented to the hospital with initial symptoms of carcinoid syndrome that later evolved into symptoms of hyperinsulinemic hypoglycaemia. Investigations revealed a metastatic neuroendocrine tumour (NET), co-secreting both serotonin and insulin. Management involved a multimodal approach in an attempt to reduce tumour burden and achieve euglycaemia, which proved to be a significant challenge in the face of refractory hypoglycaemia despite the administration of multiple prohyperglycaemic agents in combination. Unfortunately, given the burden of metastatic disease and multiple medical complications that ensued, the patient passed away. This case highlights the clinical history of a NET co-secreting serotonin and insulin, the use of combination therapy in the treatment of refractory hypoglycaemia in a metastatic insulin-producing tumour and emerging therapeutic modalities in the treatment of these rare malignancies.


Asunto(s)
Manejo de la Enfermedad , Hipoglucemia/terapia , Insulina/biosíntesis , Tumores Neuroendocrinos/complicaciones , Octreótido/uso terapéutico , Neoplasias Pancreáticas/complicaciones , Serotonina/biosíntesis , Adulto , Biopsia , Fármacos Gastrointestinales/uso terapéutico , Humanos , Hipoglucemia/diagnóstico , Hipoglucemia/etiología , Masculino , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/metabolismo , Tomografía de Emisión de Positrones , Tomografía Computarizada por Rayos X
15.
J Diabetes Res ; 2020: 5740923, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33102605

RESUMEN

BACKGROUND: The long-term insulin therapy for type 1 diabetes mellitus (T1DM) fails to achieve optimal glycemic control and avoid adverse events simultaneously. Stem cells have unique immunomodulatory capacities and have been considered as a promising interventional strategy for T1DM. Stem cell therapy in T1DM has been tried in many studies. However, the results were controversial. We thus performed a meta-analysis to update the efficacy and safety of stem cell therapy in patients with T1DM. METHODS: We systematically searched the Medline, EMBASE, Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, Web of Science, Wan Fang Data, China National Knowledge Infrastructure, VIP database, and the Chinese Biomedical Literature Database (SinoMed) for relevant studies published before March 19, 2019. The outcomes included parameters for glycemic control (i.e., glycosylated hemoglobin (HbA1c) levels and insulin dosages), ß cell function (i.e., fasting C-peptide levels and area-under-curve of C-peptide concentration (AUCC)), and relative risk of adverse events. Statistical analysis was conducted by using RevMan 5.3 and Stata 12.0. RESULTS: Five randomized controlled trials (RCTs) and eight nonrandomized concurrent control trials (NRCCTs) with a total of 396 individuals were finally included into the meta-analysis. Among RCTs, stem cell therapy could significantly reduce HbA1c levels (MD = -1.20, 95% CI -1.91 to -0.49, P = 0.0009) and increase fasting C-peptide levels (MD = 0.25, 95% CI 0.04 to 0.45, P = 0.02) and AUCC (SMD = 0.66, 95% CI 0.13 to 1.18, P = 0.01). Stem cell therapy could also reduce insulin dosages (SMD = -2.65, 95% CI -4.86 to -0.45, P = 0.02) at 6 months after treatment. NRCCTs also had consistent results. Furthermore, RCTs showed stem cell therapy did not increase relative risk of gastrointestinal symptom (RR = 0.69, 95% CI 0.14 to 3.28, P = 0.64) and infection (RR = 0.97, 95% CI 0.40 to 2.34, P = 0.95). However, NRCCTs showed stem cell therapy increased relative risk of gastrointestinal symptom (RR = 44.49, 95% CI 9.20 to 215.18, P < 0.00001). CONCLUSION: Stem cell therapy for T1DM may improve glycemic control and ß cell function without increasing the risk of serious adverse events. Stem cell therapy may also have a short-term (3-6 months) effect on reducing insulin dosages.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Trasplante de Células Madre/métodos , Área Bajo la Curva , Péptido C/sangre , Enfermedades Gastrointestinales/complicaciones , Hemoglobina Glucada/biosíntesis , Humanos , Hipoglucemiantes/efectos adversos , Insulina/biosíntesis , Insulina/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Riesgo , Resultado del Tratamiento
16.
PLoS One ; 15(5): e0233502, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32433667

RESUMEN

The environment within the Endoplasmic Reticulum (ER) influences Insulin biogenesis. In particular, ER stress may contribute to the development of Type 2 Diabetes (T2D) and Cystic Fibrosis Related Diabetes (CFRD), where evidence of impaired Insulin processing, including elevated secreted Proinsulin/Insulin ratios, are observed. Our group has established the role of a novel ER chaperone ERp29 (ER protein of 29 kDa) in the biogenesis of the Epithelial Sodium Channel, ENaC. The biogenesis of Insulin and ENaC share may key features, including their potential association with COP II machinery, their cleavage into a more active form in the Golgi or later compartments, and their ability to bypass such cleavage and remain in a less active form. Given these similarities we hypothesized that ERp29 is a critical factor in promoting the efficient conversion of Proinsulin to Insulin. Here, we confirmed that Proinsulin associates with the COP II vesicle cargo recognition component, Sec24D. When Sec24D expression was decreased, we observed a corresponding decrease in whole cell Proinsulin levels. In addition, we found that Sec24D associates with ERp29 in co-precipitation experiments and that ERp29 associates with Proinsulin in co-precipitation experiments. When ERp29 was overexpressed, a corresponding increase in whole cell Proinsulin levels was observed, while depletion of ERp29 decreased whole cell Proinsulin levels. Together, these data suggest a potential role for ERp29 in regulating Insulin biosynthesis, perhaps in promoting the exit of Proinsulin from the ER via Sec24D/COPII vesicles.


Asunto(s)
Proteínas de Choque Térmico/fisiología , Insulina/biosíntesis , Proteínas de Transporte Vesicular/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Fibrosis Quística/complicaciones , Diabetes Mellitus Tipo 2 , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Humanos , Proinsulina/metabolismo , Transporte de Proteínas
17.
BMC Med Genet ; 21(1): 91, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375679

RESUMEN

BACKGROUND: Renal hypouricemia (RHUC) is a hereditary disorder where mutations in SLC22A12 gene and SLC2A9 gene cause RHUC type 1 (RHUC1) and RHUC type 2 (RHUC2), respectively. These genes regulate renal tubular reabsorption of urates while there exist other genes counterbalancing the net excretion of urates including ABCG2 and SLC17A1. Urate metabolism is tightly interconnected with glucose metabolism, and SLC2A9 gene may be involved in insulin secretion from pancreatic ß-cells. On the other hand, a myriad of genes are responsible for the impaired insulin secretion independently of urate metabolism. CASE PRESENTATION: We describe a 67 year-old Japanese man who manifested severe hypouricemia (0.7 mg/dl (3.8-7.0 mg/dl), 41.6 µmol/l (226-416 µmol/l)) and diabetes with impaired insulin secretion. His high urinary fractional excretion of urate (65.5%) and low urinary C-peptide excretion (25.7 µg/day) were compatible with the diagnosis of RHUC and impaired insulin secretion, respectively. Considering the fact that metabolic pathways regulating urates and glucose are closely interconnected, we attempted to delineate the genetic basis of the hypouricemia and the insulin secretion defect observed in this patient using whole exome sequencing. Intriguingly, we found homozygous Trp258* mutations in SLC22A12 gene causing RHUC1 while concurrent mutations reported to be associated with hyperuricemia were also discovered including ABCG2 (Gln141Lys) and SLC17A1 (Thr269Ile). SLC2A9, that also facilitates glucose transport, has been implicated to enhance insulin secretion, however, the non-synonymous mutations found in SLC2A9 gene of this patient were not dysfunctional variants. Therefore, we embarked on a search for causal mutations for his impaired insulin secretion, resulting in identification of multiple mutations in HNF1A gene (MODY3) as well as other genes that play roles in pancreatic ß-cells. Among them, the Leu80fs in the homeobox gene NKX6.1 was an unreported mutation. CONCLUSION: We found a case of RHUC1 carrying mutations in SLC22A12 gene accompanied with compensatory mutations associated with hyperuricemia, representing the first report showing coexistence of the mutations with opposed potential to regulate urate concentrations. On the other hand, independent gene mutations may be responsible for his impaired insulin secretion, which contains novel mutations in key genes in the pancreatic ß-cell functions that deserve further scrutiny.


Asunto(s)
Complicaciones de la Diabetes/genética , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Transportadores de Anión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/genética , Defectos Congénitos del Transporte Tubular Renal/genética , Cálculos Urinarios/genética , Anciano , Complicaciones de la Diabetes/complicaciones , Complicaciones de la Diabetes/patología , Glucosa/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Heterocigoto , Proteínas de Homeodominio/genética , Homocigoto , Humanos , Insulina/biosíntesis , Insulina/genética , Secreción de Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Mutación/genética , Defectos Congénitos del Transporte Tubular Renal/complicaciones , Defectos Congénitos del Transporte Tubular Renal/patología , Ácido Úrico/metabolismo , Cálculos Urinarios/complicaciones , Cálculos Urinarios/patología , Secuenciación del Exoma
18.
J Biochem Mol Toxicol ; 34(7): e22488, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32128977

RESUMEN

Insulinomas originate from pancreatic ß cells and it is the most widely known tumor. Indomethacin is a nonsteroidal anti-inflammatory drug, which is used for blocking the production of some natural substances that cause inflammation and decrease pain. In this study, I aimed to investigate the effects of indomethacin on rat insulinoma INS-1 cells. The relationship between cell death and insulin metabolism was determined with the administration of indomethacin. The cell viability by WST-1; the apoptosis and necrosis levels by ELISA kits; malondialdehyde levels by spectrophotometer; and beclin, intracellular insulin, insulin secretion, KCa3.1, insulin receptor (IR), glucose transporter type 2 (GLUT2), activating transcription factor 2 (ATF2), Elk1, c-Jun, Akt and phosphorylated ATF2, Elk1, c-Jun, Akt, intracellular betacellulin and betacellulin secretion levels by Western blot analysis investigated. The Ins1, Ins2, IR, GLUT2, ATF2, Elk1, c-Jun, Akt, and Betacellulin gene expression levels were determined by the real-time quantitative reverse transcription-polymerase chain reaction method. Apoptotic cell death was observed with the administration of indomethacin. The insulin secretion and Ins1, Ins2 gene expression levels decreased. The insulin receptor and GLUT2 levels increased, while KCa3.1 (KCNN4) levels decreased with the administration of indomethacin to insulinoma INS-1 cells. A decrease was observed in the total c-Jun, phosphorylated ATF2, Elk1, c-Jun, and Akt levels. Betacellulin secretion levels increased. In insulinoma INS-1 cells, apoptotic cell death occurred in the following manner: (i) indomethacin might decrease insulin secretion by reducing KCa3.1, (ii) might inactivate the JNK/ERK pathway with the inactivity of transcription factors.


Asunto(s)
Apoptosis/efectos de los fármacos , Indometacina/farmacología , Secreción de Insulina/efectos de los fármacos , Insulina/biosíntesis , Insulinoma/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias Pancreáticas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/patología , Neoplasias Pancreáticas/patología , Ratas
19.
Differentiation ; 113: 1-9, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32120156

RESUMEN

The functional maturation of human pancreatic ß-cells remains poorly understood. EndoC-ßH2 is a human ß-cell line with a reversible immortalized phenotype. Removal of the two oncogenes, SV40LT and hTERT introduced for its propagation, stops proliferation, triggers cell size increase and senescence, promotes mitochondrial activity and amplifies several ß-cell traits and functions. Overall, these events recapitulate several aspects of functional ß-cell maturation. We report here that selective depletion of SV40LT, but not of hTERT, is sufficient to revert EndoC-ßH2 immortalization. SV40LT inhibits the activity of the RB family members and of P53. In EndoC-ßH2 cells, the knock-down of RB itself, and, to a lesser extent, of its relative P130, precludes most events triggered by SV40LT depletion. In contrast, the knock-down of P53 does not prevent reversion of immortalization. Thus, an increase in RB and P130 activity, but not in P53 activity, is required for functional maturation of EndoC-ßH2 cells upon SV40LT-depletion. In addition, RB and/or P130 depletion in SV40LT-expressing EndoC-ßH2 cells decreases cell size, stimulates proliferation, and decreases the expression of key ß-cell genes. Thus, despite SV40LT expression, EndoC-ßH2 cells have a residual RB activity, which when suppressed reverts them to a more immature phenotype. These results show that the expression and activity levels of RB family members, especially RB itself, regulate the maturation state of EndoC-ßH2 cells.


Asunto(s)
Genes de Retinoblastoma , Células Secretoras de Insulina/metabolismo , Proteína de Retinoblastoma/fisiología , Antígenos Transformadores de Poliomavirus/genética , Ciclo Celular , Línea Celular , Proliferación Celular , Senescencia Celular , Técnicas de Silenciamiento del Gen , Humanos , Insulina/biosíntesis , Insulina/genética , Células Secretoras de Insulina/citología , Familia de Multigenes , ARN Interferente Pequeño , Proteína p130 Similar a la del Retinoblastoma/fisiología , Telomerasa/genética , Transcripción Genética , Proteína p53 Supresora de Tumor/fisiología
20.
Biomed Res Int ; 2020: 7103053, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32051828

RESUMEN

Mesenchymal stem cells (MSCs) can be differentiated in vitro to form insulin-producing cells (IPCs). However, the proportion of induced cells is modest. Extracts from injured pancreata of rodents promoted this differentiation, and three upregulated proteins were identified in these extracts. The aim of this study was to evaluate the potential benefits of adding these proteins to the differentiation medium alone or in combination. Our results indicate that the proportion of IPCs among the protein(s)-supplemented samples was significantly higher than that in the samples with no added proteins. The yield from samples supplemented with PRDX6 alone was 4-fold higher than that from samples without added protein. These findings were also supported by the results of fluorophotometry. Gene expression profiles revealed higher levels among protein-supplemented samples. Significantly higher levels of GGT, SST, Glut-2, and MafB expression were noted among PRDX6-treated samples. There was a stepwise increase in the release of insulin and c-peptide, as a function of increasing glucose concentrations, indicating that the differentiated cells were glucose sensitive and insulin responsive. PRDX6 exerts its beneficial effects as a result of its biological antioxidant properties. Considering its ease of use as a single protein, PRDX6 is now routinely used in our differentiation protocols.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Insulina/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Peroxiredoxina VI/metabolismo , Peroxiredoxina VI/farmacología , Péptido C/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Humanos , Factor de Transcripción MafB/metabolismo , Peroxiredoxina VI/genética , Somatostatina/metabolismo , Transcriptoma , gamma-Glutamiltransferasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA