Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 510
Filtrar
1.
Cell Transplant ; 30: 963689721999330, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33902338

RESUMO

Circulating microRNAs (miRNAs) can be biomarkers for diagnosis and progression of several pathophysiological conditions. In a cohort undergoing total pancreatectomy with islet autotransplantation (TPIAT) from the multicenter Prospective Observational Study of TPIAT (POST), we investigated associations between a panel of circulating miRNAs (hsa-miR-375, hsa-miR-29b-3p, hsa-miR-148a-3p, hsa-miR-216a-5p, hsa-miR-320d, hsa-miR-200c, hsa-miR-125b, hsa-miR-7-5p, hsa-miR-221-3p, hsa-miR-122-5p) and patient, disease and islet-isolation characteristics. Plasma samples (n = 139) were collected before TPIAT and miRNA levels were measured by RTPCR. Disease duration, prior surgery, and pre-surgical diabetes were not associated with circulating miRNAs. Levels of hsa-miR-29b-3p (P = 0.03), hsa-miR-148a-3p (P = 0.04) and hsa-miR-221-3p (P = 0.01) were lower in those with genetic risk factors. Levels of hsa-miR-148a-3p (P = 0.04) and hsa-miR-7-5p (P = 0.04) were elevated in toxic/metabolic disease. Participants with exocrine insufficiency had lower hsa-miR-29b-3p, hsa-miR-148a-3p, hsa-miR-320d, hsa-miR-221-3p (P < 0.01) and hsa-miR-375, hsa-miR-200c-3p, and hsa-miR-125b-5p (P < 0.05). Four miRNAs were associated with fasting C-peptide before TPIAT (hsa-miR-29b-3p, r = 0.18; hsa-miR-148a-3p, r = 0.21; hsa-miR-320d, r = 0.19; and hsa-miR-221-3p, r = 0.21; all P < 0.05), while hsa-miR-29b-3p was inversely associated with post-isolation islet equivalents/kg and islet number/kg (r = -0.20, P = 0.02). Also, hsa-miR-200c (r = 0.18, P = 0.03) and hsa-miR-221-3p (r = 0.19, P = 0.03) were associated with islet graft tissue volume. Further investigation is needed to determine the predictive potential of these miRNAs for assessing islet autotransplant outcomes.


Assuntos
Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/fisiopatologia , MicroRNAs/metabolismo , Pancreatectomia/métodos , Transplante Autólogo/métodos , Adulto , Feminino , Humanos , Masculino , Estudos Prospectivos
2.
Diabetologia ; 64(3): 630-640, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33404684

RESUMO

AIMS/HYPOTHESIS: Congenital hyperinsulinism caused by mutations in the KATP-channel-encoding genes (KATPHI) is a potentially life-threatening disorder of the pancreatic beta cells. No optimal medical treatment is available for patients with diazoxide-unresponsive diffuse KATPHI. Therefore, we aimed to create a model of KATPHI using patient induced pluripotent stem cell (iPSC)-derived islets. METHODS: We derived iPSCs from a patient carrying a homozygous ABCC8V187D mutation, which inactivates the sulfonylurea receptor 1 (SUR1) subunit of the KATP-channel. CRISPR-Cas9 mutation-corrected iPSCs were used as controls. Both were differentiated to stem cell-derived islet-like clusters (SC-islets) and implanted into NOD-SCID gamma mice. RESULTS: SUR1-mutant and -corrected iPSC lines both differentiated towards the endocrine lineage, but SUR1-mutant stem cells generated 32% more beta-like cells (SC-beta cells) (64.6% vs 49.0%, p = 0.02) and 26% fewer alpha-like cells (16.1% vs 21.8% p = 0.01). SUR1-mutant SC-beta cells were 61% more proliferative (1.23% vs 0.76%, p = 0.006), and this phenotype could be induced in SUR1-corrected cells with pharmacological KATP-channel inactivation. The SUR1-mutant SC-islets secreted 3.2-fold more insulin in low glucose conditions (0.0174% vs 0.0054%/min, p = 0.0021) and did not respond to KATP-channel-acting drugs in vitro. Mice carrying grafts of SUR1-mutant SC-islets presented with 38% lower fasting blood glucose (4.8 vs 7.7 mmol/l, p = 0.009) and their grafts failed to efficiently shut down insulin secretion during induced hypoglycaemia. Explanted SUR1-mutant grafts displayed an increase in SC-beta cell proportion and SC-beta cell nucleomegaly, which was independent of proliferation. CONCLUSIONS/INTERPRETATION: We have created a model recapitulating the known pathophysiology of KATPHI both in vitro and in vivo. We have also identified a novel role for KATP-channel activity during human islet development. This model will enable further studies for the improved understanding and clinical management of KATPHI without the need for primary patient tissue.


Assuntos
Hiperinsulinismo Congênito/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores de Sulfonilureias/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Hiperinsulinismo Congênito/genética , Hiperinsulinismo Congênito/patologia , Hiperinsulinismo Congênito/fisiopatologia , Feminino , Predisposição Genética para Doença , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/transplante , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação , Fenótipo , Receptores de Sulfonilureias/genética
3.
Diabetologia ; 63(10): 1966-1973, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32894306

RESUMO

For much of the last century, our knowledge regarding the pancreas in type 1 and type 2 diabetes was largely derived from autopsy studies of individuals with these disorders or investigations utilising rodent models of either disease. While many important insights emanated from these efforts, the mode for investigation has increasingly seen change due to the availability of transplant-quality organ-donor tissues, improvements in pancreatic imaging, advances in metabolic assessments of living patients, genetic analyses, technological advances for laboratory investigation and more. As a result, many long-standing notions regarding the role for and the changes that occur in the pancreas in individuals with these disorders have come under question, while, at the same time, new issues (e.g., beta cell persistence, disease heterogeneity, exocrine contributions) have arisen. In this article, we will consider the vital role of the pancreas in human health and physiology, including discussion of its anatomical features and dual (exocrine and endocrine) functions. Specifically, we convey changes that occur in the pancreas of those with either type 1 or type 2 diabetes, with careful attention to the facets that may contribute to the pathogenesis of either disorder. Finally, we discuss the emerging unknowns with the belief that understanding the role of the pancreas in type 1 and type 2 diabetes will lead to improvements in disease diagnosis, understanding of disease heterogeneity and optimisation of treatments at a personalised level. Graphical abstract.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Pâncreas/metabolismo , Tecido Adiposo/patologia , Amiloidose/metabolismo , Amiloidose/patologia , Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/imunologia , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Pâncreas/patologia , Pâncreas/fisiopatologia , Pâncreas Exócrino/metabolismo , Pâncreas Exócrino/patologia , Pâncreas Exócrino/fisiopatologia , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/patologia
4.
Diabetologia ; 63(10): 2030-2039, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32894313

RESUMO

The islets of Langerhans are well embedded within the exocrine pancreas (the latter comprised of ducts and acini), but the nature of interactions between these pancreatic compartments and their role in determining normal islet function and survival are poorly understood. However, these interactions appear to be critical, as when pancreatic exocrine disease occurs, islet function and insulin secretion frequently decline to the point that diabetes ensues, termed pancreatogenic diabetes. The most common forms of pancreatogenic diabetes involve sustained exocrine disease leading to ductal obstruction, acinar inflammation, and fibro-fatty replacement of the exocrine pancreas that predates the development of dysfunction of the endocrine pancreas, as seen in chronic pancreatitis-associated diabetes and cystic fibrosis-related diabetes and, more rarely, MODY type 8. Intriguingly, a form of tumour-induced diabetes has been described that is associated with pancreatic ductal adenocarcinoma. Here, we review the similarities and differences among these forms of pancreatogenic diabetes, with the goal of highlighting the importance of exocrine/ductal homeostasis for the maintenance of pancreatic islet function and survival and to highlight the need for a better understanding of the mechanisms underlying these diverse conditions. Graphical abstract.


Assuntos
Fibrose Cística/metabolismo , Diabetes Mellitus/metabolismo , Ilhotas Pancreáticas/metabolismo , Pâncreas Exócrino/metabolismo , Pancreatite Crônica/metabolismo , Animais , Carcinoma Ductal Pancreático/complicações , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Fibrose Cística/complicações , Fibrose Cística/patologia , Diabetes Mellitus/etiologia , Humanos , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Pâncreas Exócrino/patologia , Pâncreas Exócrino/fisiopatologia , Pancreatopatias/complicações , Pancreatopatias/metabolismo , Pancreatopatias/patologia , Neoplasias Pancreáticas/complicações , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Pancreatite Crônica/complicações , Pancreatite Crônica/patologia
5.
Surgery ; 168(6): 1032-1040, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32843212

RESUMO

BACKGROUND: Necrotizing pancreatitis survivors develop complications beyond infected necrosis that often require invasive intervention. Remarkably few data have cataloged these late complications after acute necrotizing pancreatitis resolution. We sought to identify the types and incidence of complications after necrotizing pancreatitis. DESIGN: An observational study was performed evaluating 647 patients with necrotizing pancreatitis captured in a single-institution database between 2005 and 2017 at a tertiary care hospital. Retrospective review and analysis of newly diagnosed conditions attributable to necrotizing pancreatitis was performed. Exclusion criteria included the following: death before disease resolution (n = 57, 9%) and patients lost to follow-up (n = 12, 2%). RESULTS: A total of 578 patients were followed for a median of 46 months (range, 8 months to 15 y) after necrotizing pancreatitis. In 489 (85%) patients 1 or more complications developed and included symptomatic disconnected pancreatic duct syndrome (285 of 578, 49%), splanchnic vein thrombosis (257 of 572, 45%), new endocrine insufficiency (195 of 549, 35%), new exocrine insufficiency (108 of 571, 19%), symptomatic chronic pancreatitis (93 of 571, 16%), incisional hernia (89 of 420, 21%), biliary stricture (90 of 576, 16%), chronic pain (44 of 575, 8%), gastrointestinal fistula (44 of 578, 8%), pancreatic duct stricture (30 of 578, 5%), and duodenal stricture (28 of 578, 5%). During the follow-up period, a total of 340 (59%) patients required an invasive intervention after necrotizing pancreatitis resolution. Invasive pancreatobiliary intervention was required in 230 (40%) patients. CONCLUSION: Late complications are common in necrotizing pancreatitis survivors. A broad variety of problems manifest themselves after resolution of the acute disease process and often require invasive intervention. Necrotizing pancreatitis patients should be followed lifelong by experienced clinicians.


Assuntos
Pancreatite Necrosante Aguda/complicações , Sobreviventes/estatística & dados numéricos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Dor Crônica/epidemiologia , Dor Crônica/etiologia , Constrição Patológica/epidemiologia , Constrição Patológica/etiologia , Drenagem/efeitos adversos , Insuficiência Pancreática Exócrina/epidemiologia , Insuficiência Pancreática Exócrina/etiologia , Feminino , Seguimentos , Fístula Gástrica/epidemiologia , Fístula Gástrica/etiologia , Humanos , Incidência , Fístula Intestinal/epidemiologia , Fístula Intestinal/etiologia , Ilhotas Pancreáticas/fisiopatologia , Masculino , Pessoa de Meia-Idade , Pancreatectomia/efeitos adversos , Pancreatite Necrosante Aguda/mortalidade , Pancreatite Necrosante Aguda/fisiopatologia , Pancreatite Necrosante Aguda/terapia , Pancreatite Crônica/epidemiologia , Pancreatite Crônica/etiologia , Estudos Retrospectivos , Circulação Esplâncnica , Trombose Venosa/epidemiologia , Trombose Venosa/etiologia , Adulto Jovem
6.
J Clin Endocrinol Metab ; 105(10)2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32668452

RESUMO

CONTEXT: Oral glucose tolerance test (OGTT)-related hypoglycemia is common in pancreatic-insufficient cystic fibrosis (PI-CF), but its mechanistic underpinnings are yet to be established. OBJECTIVE: To delineate the mechanism(s) underlying OGTT-related hypoglycemia. DESIGN AND SETTING: We performed 180-minute OGTTs with frequent blood sampling in adolescents and young adults with PI-CF and compared results with those from a historical healthy control group. Hypoglycemia (Hypo[+]) was defined as plasma glucose <65 mg/dL. We hypothesized that CF-Hypo[+] would demonstrate impaired early phase insulin secretion and persistent late insulin effect compared with control-Hypo[+], and explored the contextual counterregulatory response. MAIN OUTCOME MEASURE: OGTT 1-hour and nadir glucose, insulin, C-peptide, and insulin secretory rate (ISR) incremental areas under the curve (AUC) between 0 and 30 minutes (early) and between 120 and 180 minutes (late), and Δglucagon120-180min and Δfree fatty acids (FFAs)120-180min were compared between individuals with CF and control participants with Hypo[+]. RESULTS: Hypoglycemia occurred in 15/23 (65%) patients with CF (43% female, aged 24.8 [14.6-30.6] years) and 8/15 (55%) control participants (33% female, aged 26 [21-38] years). The CF-Hypo[+] group versus the control-Hypo[+] group had higher 1-hour glucose (197 ± 49 vs 139 ± 53 mg/dL; P = 0.05) and lower nadir glucose levels (48 ± 7 vs 59 ± 4 mg/dL; P < 0.01), while insulin, C-peptide, and ISR-AUC0-30 min results were lower and insulin and C-peptide, and AUC120-180min results were higher (P < 0.05). Individuals with CF-Hypo[+] had lower Δglucagon120-180min and ΔFFA120-180min compared with the control-Hypo[+] group (P < 0.01). CONCLUSIONS: OGTT-related hypoglycemia in PI-CF is associated with elevated 1-hour glucose, impaired early phase insulin secretion, higher late insulin exposure, and less increase in glucagon and FFAs. These data suggest that hypoglycemia in CF is a manifestation of islet dysfunction including an impaired counterregulatory response.


Assuntos
Fibrose Cística/complicações , Hipoglicemia/fisiopatologia , Secreção de Insulina/fisiologia , Ilhotas Pancreáticas/fisiopatologia , Adolescente , Adulto , Glicemia/análise , Peptídeo C/sangue , Estudos de Casos e Controles , Criança , Fibrose Cística/sangue , Fibrose Cística/fisiopatologia , Feminino , Glucagon/sangue , Teste de Tolerância a Glucose/métodos , Humanos , Hipoglicemia/sangue , Hipoglicemia/diagnóstico , Hipoglicemia/etiologia , Insulina/sangue , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Fatores de Tempo , Adulto Jovem
7.
Cells ; 9(3)2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32168890

RESUMO

Insulin secretion from the ß-cells of the islets of Langerhans is triggered mainly by nutrients such as glucose, and incretin hormones such as glucagon-like peptide-1 (GLP-1). The mechanisms of the stimulus-secretion coupling involve the participation of the key enzymes that metabolize the nutrients, and numerous ion channels that mediate the electrical activity. Several members of the transient receptor potential (TRP) channels participate in the processes that mediate the electrical activities and Ca2+ oscillations in these cells. Human ß-cells express TRPC1, TRPM2, TRPM3, TRPM4, TRPM7, TRPP1, TRPML1, and TRPML3 channels. Some of these channels have been reported to mediate background depolarizing currents, store-operated Ca2+ entry (SOCE), electrical activity, Ca2+ oscillations, gene transcription, cell-death, and insulin secretion in response to stimulation by glucose and GLP1. Different channels of the TRP family are regulated by one or more of the following mechanisms: activation of G protein-coupled receptors, the filling state of the endoplasmic reticulum Ca2+ store, heat, oxidative stress, or some second messengers. This review briefly compiles our current knowledge about the molecular mechanisms of regulations, and functions of the TRP channels in the ß-cells, the α-cells, and some insulinoma cell lines.


Assuntos
Insulinoma/fisiopatologia , Ilhotas Pancreáticas/fisiopatologia , Canais de Potencial de Receptor Transitório/metabolismo , Humanos
8.
Arch Physiol Biochem ; 126(3): 235-241, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30293453

RESUMO

Islet dysfunction is a hallmark of type 2 diabetes mellitus (T2DM). Compelling evidence suggests that accumulation of islet amyloid in the islets of Langerhans significantly contribute to ß-cell dysfunction and diabetes. Emerging evidence implicates a role for cystic fibrosis transmembrane-conductance regulator in the regulation of insulin secretion from pancreatic islets. Impaired first-phase insulin responses and glucose homeostasis have also been reported in cystic fibrosis patients. The transforming growth factor-ß protein superfamily is central regulators of pancreatic cell function, and has a key role in pancreas development and pancreatic disease, including diabetes and islet dysfunction. It is also becoming clear that islet inflammation plays a key role in the development of islet dysfunction. Inflammatory changes, including accumulation of macrophages, have been documented in type 2 diabetic islets. Islet dysfunction leads to hyperglycemia and ultimately the development of diabetes. In this review, we describe these risk factors and their associations with islet dysfunction.


Assuntos
Fibrose Cística/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Ilhotas Pancreáticas/citologia , Amiloide/metabolismo , Animais , Fibrose Cística/complicações , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diabetes Mellitus Tipo 2/complicações , Progressão da Doença , Glucose/metabolismo , Homeostase , Humanos , Hiperglicemia , Inflamação , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/fisiopatologia , Macrófagos/metabolismo , Pâncreas/fisiopatologia , Fatores de Risco , Fator de Crescimento Transformador beta/metabolismo
9.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31677389

RESUMO

CONTEXT: Chronic inflammation arising from adipose tissue macrophage (ATM) activation may be central in type 2 diabetes etiology. Our objective was to assess the longitudinal associations of soluble CD163 (sCD163), a novel biomarker of ATM activation, with insulin sensitivity, ß-cell function, and dysglycemia in high-risk subjects. METHODS: Adults at risk for type 2 diabetes in the Prospective Metabolism and Islet Cell Evaluation (PROMISE) study had 3 assessments over 6 years (n = 408). Levels of sCD163 were measured using fasting serum. Insulin sensitivity was assessed by HOMA2-%S and the Matsuda index (ISI). ß-cell function was determined by insulinogenic index (IGI) over HOMA-IR and insulin secretion-sensitivity index-2 (ISSI-2). Incident dysglycemia was defined as the onset of impaired fasting glucose, impaired glucose tolerance, or type 2 diabetes. Generalized estimating equations (GEE) evaluated longitudinal associations of sCD163 with insulin sensitivity, ß-cell function, and incident dysglycemia adjusting for demographic and lifestyle covariates. Areas under receiver-operating-characteristic curve (AROC) tested whether sCD163 improved dysglycemia prediction in a clinical model. RESULTS: Longitudinal analyses showed significant inverse associations between sCD163 and insulin sensitivity (% difference per standard deviation increase of sCD163 for HOMA2-%S (ß = -7.01; 95% CI, -12.26 to -1.44) and ISI (ß = -7.60; 95% CI, -11.09 to -3.97) and ß-cell function (ISSI-2 (ß = -4.67; 95 %CI, -8.59 to -0.58) and IGI/HOMA-IR (ß = -8.75; 95% CI, -15.42 to -1.56)). Increased sCD163 was associated with greater risk for incident dysglycemia (odds ratio = 1.04; 95% CI, 1.02-1.06; P < 0.001). Adding sCD163 data to a model with clinical variables improved prediction of incident dysglycemia (AROC=0.6731 vs 0.638; P < 0.05). CONCLUSIONS: sCD163 was longitudinally associated with core disorders that precede the onset of type 2 diabetes.


Assuntos
Antígenos CD/sangue , Antígenos de Diferenciação Mielomonocítica/sangue , Diabetes Mellitus Tipo 2/imunologia , Resistência à Insulina/imunologia , Ativação de Macrófagos , Macrófagos/metabolismo , Receptores de Superfície Celular/sangue , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Biomarcadores/sangue , Biomarcadores/metabolismo , Glicemia/análise , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/fisiopatologia , Feminino , Teste de Tolerância a Glucose , Humanos , Ilhotas Pancreáticas/fisiopatologia , Estudos Longitudinais , Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Receptores de Superfície Celular/metabolismo
10.
Pancreas ; 48(9): 1182-1187, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31593011

RESUMO

OBJECTIVE: The aim of the study was to evaluate the long-term functional outcome (exocrine and endocrine) and morphological changes in remnant pancreas after pancreatoduodenectomy and its clinical impact. METHODS: Periampullary carcinoma patients with minimum follow-up of 2 years and without recurrence were included (N = 102). Exocrine insufficiency includes clinical steatorrhea and fecal elastase-1 (FE-1) levels; endocrine insufficiency, glucose levels and glycated hemoglobin; and morphological changes, main pancreatic duct (MPD) diameter and thickness of remnant pancreas. RESULTS: The mean (standard deviation) follow-up period was 59 (26) months. Of the 102 patients, 81 (80%) had severely deficient FE-1 (0-100 µg/g). The preoperative MPD was significantly more and thickness of remnant pancreas was significantly less in patients with severely deficient FE-1. Overall, 15.6% (16/102) developed steatorrhea and improved on enzyme replacement therapy. The presence of MPD stricture (P = 0.008) and weight loss (P = 0.001) were significantly associated with steatorrhea. New-onset diabetes was seen in 17% (15/90) patients, of whom 3 of 5 developed it after 4 years (range, 4-7 years). The blood glucose was controlled on oral hypoglycemics in 2 (10/15) of 3 patients. CONCLUSIONS: The assessment by FE-1 indicates loss of exocrine function in more than 90%, whereas only 1 of 6 developed steatorrhea and new-onset diabetes. Morphological changes especially MPD stricture affect the functional status of remnant pancreas.


Assuntos
Ampola Hepatopancreática/cirurgia , Carcinoma/cirurgia , Neoplasias do Ducto Colédoco/cirurgia , Avaliação de Resultados em Cuidados de Saúde/estatística & dados numéricos , Pancreaticoduodenectomia/métodos , Adulto , Ampola Hepatopancreática/patologia , Insuficiência Pancreática Exócrina/diagnóstico , Insuficiência Pancreática Exócrina/fisiopatologia , Fezes/enzimologia , Feminino , Seguimentos , Humanos , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde/métodos , Pâncreas/patologia , Pâncreas/fisiopatologia , Pâncreas Exócrino/patologia , Pâncreas Exócrino/fisiopatologia , Ductos Pancreáticos/patologia , Ductos Pancreáticos/fisiopatologia , Elastase Pancreática/metabolismo , Esteatorreia/diagnóstico , Esteatorreia/fisiopatologia , Fatores de Tempo
11.
Pancreas ; 48(9): 1160-1166, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31593013

RESUMO

OBJECTIVE: Little data exist describing the change over time in islet function and glycemic control in patients with chronic pancreatitis (CP). METHODS: In 325 CP patients who underwent 2 mixed meal tolerance tests and/or glycated hemoglobin (HbA1c) levels, we estimated the rate of change in metabolic measures per 6 months and assessed the association between potential risk factors for diabetes and rate of change using multivariate regression models. RESULTS: Per 6-month time, HbA1c increased by 0.062% with a standard error of 0.029% (P = 0.037) and the ratio (area under the curve (AUC) C-peptide to AUC glucose from mixed meal tolerance testing) decreased by 0.0028 with a standard error of 0.0011 (P = 0.014). We observed more rapid decline in smokers (AUC C-peptide, P = 0.043) and patients with surgical drainage (AUC glucose, P = 0.001; ratio, P = 0.03) or with calcific pancreatitis (HbA1c, P = 0.003). In multivariate models, AUC C-peptide and ratio declined at a greater rate in smokers and HbA1c in those with pancreatic calcifications (both P < 0.05). CONCLUSIONS: We observed a measurable decline in ß-cell function and glycemic control in patients with CP. Patients with a history of tobacco smoking, surgical drainage, or pancreatic calcification may be at highest risk.


Assuntos
Glicemia/metabolismo , Peptídeo C/sangue , Hemoglobinas Glicadas/metabolismo , Ilhotas Pancreáticas/fisiopatologia , Pancreatopatias/fisiopatologia , Pancreatite Crônica/fisiopatologia , Adolescente , Adulto , Diabetes Mellitus/sangue , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Progressão da Doença , Feminino , Teste de Tolerância a Glucose , Humanos , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Masculino , Pessoa de Meia-Idade , Pancreatopatias/metabolismo , Pancreatite Crônica/metabolismo , Pancreatite Crônica/terapia , Fatores de Risco , Adulto Jovem
12.
J Clin Endocrinol Metab ; 104(11): 5703-5714, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31390002

RESUMO

OBJECTIVE: Augmenting nicotinamide adenine dinucleotide (NAD+) metabolism through dietary provision of NAD+ precursor vitamins translates to improved glucose handling in rodent models of obesity and diabetes. Preclinical evidence suggests that the NAD+/SIRT1 axis may be implicated in modulating important gut-related aspects of glucose regulation. We sought to test whether NAD+ precursor supplementation with nicotinamide riboside (NR) affects ß-cell function, α-cell function, and incretin hormone secretion as well as circulating bile acid levels in humans. DESIGN: A 12-week randomized, double-blind, placebo-controlled, parallel-group trial in 40 males with obesity and insulin resistance allocated to NR at 1000 mg twice daily (n = 20) or placebo (n = 20). Two-hour 75-g oral glucose tolerance tests were performed before and after the intervention, and plasma concentrations of glucose, insulin, C-peptide, glucagon, glucagon-like peptide 1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP) were determined. ß-Cell function indices were calculated based on glucose, insulin, and C-peptide measurements. Fasting plasma concentrations of bile acids were determined. RESULTS: NR supplementation during 12 weeks did not affect fasting or postglucose challenge concentrations of glucose, insulin, C-peptide, glucagon, GLP-1, or GIP, and ß-cell function did not respond to the intervention. Additionally, no changes in circulating adipsin or bile acids were observed following NR supplementation. CONCLUSION: The current study does not provide evidence to support that dietary supplementation with the NAD+ precursor NR serves to impact glucose tolerance, ß-cell secretory capacity, α-cell function, and incretin hormone secretion in nondiabetic males with obesity. Moreover, bile acid levels in plasma did not change in response to NR supplementation.


Assuntos
Glicemia , Polipeptídeo Inibidor Gástrico/sangue , Peptídeo 1 Semelhante ao Glucagon/sangue , Glucagon/sangue , Ilhotas Pancreáticas/efeitos dos fármacos , Niacinamida/análogos & derivados , Obesidade/sangue , Peptídeo C/sangue , Método Duplo-Cego , Teste de Tolerância a Glucose , Humanos , Insulina/sangue , Ilhotas Pancreáticas/fisiopatologia , Masculino , Pessoa de Meia-Idade , Niacinamida/farmacologia , Obesidade/fisiopatologia , Compostos de Piridínio
13.
Nat Commun ; 10(1): 3124, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31311920

RESUMO

Cystic fibrosis (CF) is a genetic disorder caused by defective CF Transmembrane Conductance Regulator (CFTR) function. Insulin producing pancreatic islets are located in close proximity to the pancreatic duct and there is a possibility of impaired cell-cell signaling between pancreatic ductal epithelial cells (PDECs) and islet cells as causative in CF. To study this possibility, we present an in vitro co-culturing system, pancreas-on-a-chip. Furthermore, we present an efficient method to micro dissect patient-derived human pancreatic ducts from pancreatic remnant cell pellets, followed by the isolation of PDECs. Here we show that defective CFTR function in PDECs directly reduced insulin secretion in islet cells significantly. This uniquely developed pancreatic function monitoring tool will help to study CF-related disorders in vitro, as a system to monitor cell-cell functional interaction of PDECs and pancreatic islets, characterize appropriate therapeutic measures and further our understanding of pancreatic function.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/fisiopatologia , Células Epiteliais/patologia , Ilhotas Pancreáticas/fisiopatologia , Dispositivos Lab-On-A-Chip , Adolescente , Criança , Pré-Escolar , Técnicas de Cocultura/métodos , Fibrose Cística/patologia , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais/metabolismo , Feminino , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Microdissecção , Organoides , Ductos Pancreáticos/citologia , Ductos Pancreáticos/patologia , Cultura Primária de Células/métodos
14.
PLoS One ; 13(12): e0209448, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30571730

RESUMO

AIM: The purpose of this study was to clarify whether fatty pancreas might lead to impaired pancreatic endocrine or exocrine function. MATERIAL AND METHODS: The study involved 109 participants who had undergone the glucagon stimulation test and N-benzoyl-L-tyros-p-amino benzoic acid (BT-PABA) test to assess pancreatic function as well as unenhanced abdominal computed tomography (CT). Pancreatic endocrine impairment was defined as ΔC peptide immunoreactivity less than 2 [mmol/L] in the glucagon stimulation test, and pancreatic exocrine impairment was defined as a urinary PABA excretion rate less than 70% on the BT-PABA test. We defined as the mean CT value of pancreas / CT value of spleen (P/S ratio) as a marker to assess fatty pancreas. We analyzed the association between fatty pancreas and pancreatic impairment using the logistic regression model. The odds ratio (OR) is shown per 0.1 unit. RESULTS: Pancreatic endocrine function was impaired in 33.0% of the participants, and 56.9% of those were regarded as having pancreatic exocrine impairment. The P/S ratio was significantly correlated with pancreatic endocrine impairment in univariate analysis (OR = 0.61, 95% confidence interval (CI) = 0.43-0.83, P = 0.0013) and multivariate analysis (OR = 0.38, 95% CI = 0.22-0.61, P < .0001) for all participants. Similar significant relationships were observed in both univariate (OR = 0.70, 95% CI = 0.49-0.99, P = 0.04) and multivariate (OR = 0.39, 95% CI = 0.21-0.66, P = 0.0002) analyses for the participants without diabetes (n = 93). The amount of pancreatic fat was not associated with exocrine impairment in univariate analysis (OR = 0.80, 95% CI = 0.59-1.06, P = 0.12). CONCLUSION: Fatty pancreas was associated with pancreatic endocrine impairment but did not have a clear relationship with pancreatic exocrine impairment.


Assuntos
Ilhotas Pancreáticas/fisiopatologia , Lipomatose/fisiopatologia , Pâncreas Exócrino/fisiopatologia , Pancreatopatias/fisiopatologia , Ácido 4-Aminobenzoico/urina , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Glucagon/administração & dosagem , Humanos , Gordura Intra-Abdominal/diagnóstico por imagem , Ilhotas Pancreáticas/diagnóstico por imagem , Ilhotas Pancreáticas/efeitos dos fármacos , Lipomatose/diagnóstico por imagem , Lipomatose/urina , Masculino , Pessoa de Meia-Idade , Pâncreas Exócrino/diagnóstico por imagem , Pâncreas Exócrino/efeitos dos fármacos , Pancreatopatias/diagnóstico por imagem , Pancreatopatias/urina , Baço/diagnóstico por imagem , Tomografia Computadorizada por Raios X , para-Aminobenzoatos/administração & dosagem
15.
Curr Opin Pharmacol ; 43: 59-65, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30199758

RESUMO

Pregnancy involves a progressive increase in insulin resistance and the ß-cells must adapt to compensate and prevent gestational diabetes (GDM). In this review we discuss the evidence for placental peptides, including placental lactogen, hepatocyte growth factor, adiponectin and leptin, playing a role in the islet adaptation to pregnancy. The difficulties of translating data from rodent models into human pregnancy are covered and we summarise studies investigating associations between serum placental peptides and GDM risk. In conclusion, current data support important roles for placental peptides interacting to support ß-cells during pregnancy, however mechanisms involved in humans are unclear. Further work in humans is required, but placental peptides have clinical potential from both a diagnostic and therapeutic perspective.


Assuntos
Glicemia/metabolismo , Diabetes Gestacional/metabolismo , Ilhotas Pancreáticas/metabolismo , Peptídeos/metabolismo , Placenta/metabolismo , Adaptação Fisiológica , Adipocinas/metabolismo , Animais , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Diabetes Gestacional/sangue , Diabetes Gestacional/tratamento farmacológico , Diabetes Gestacional/fisiopatologia , Feminino , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Hipoglicemiantes/uso terapêutico , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiopatologia , Peptídeos/uso terapêutico , Placenta/fisiopatologia , Lactogênio Placentário/metabolismo , Gravidez , Transdução de Sinais
16.
Food Chem Toxicol ; 120: 305-320, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30026088

RESUMO

Diabetes mellitus is characterized by hyperglycemia which causes oxidative stress. Propolis has been reported to have antihyperglycemic and antioxidant potentials. The present study therefore examined the anti-hyperglycemic, antioxidant and anti-inflammatory activities of Malaysian propolis (MP) using streptozotocin-induced diabetic rats. Ethanol extract of MP showed in vitro antioxidant (DPPH, FRAP and H2O2 radical scavenging) and α-glucosidase inhibition activities. Male Sprague Dawley rats were either treated with distilled water (normal control and diabetic control), MP (300 mg/kg b. w.), metformin (Met) (300 mg/kg b. w.) or both. After four weeks, fasting blood glucose decreased, while body weight change and serum insulin level increased significantly in MP, Met and MP + Met treated diabetic groups compared to diabetic control (DC) group. Furthermore, pancreatic antioxidant enzymes, total antioxidant capacity, interleukin (IL)-10 and proliferating cell nuclear antigen increased, while malondialdehyde, nuclear factor-kappa B (p65), tumor necrosis factor alpha, IL-1ß and cleaved caspase-3 decreased significantly in the treated diabetic groups compared to DC group. Histopathology of the pancreas showed increased islet area and number of beta cells in the treated groups, compared to DC group, with D + MP + Met group comparable to normal control. We conclude that MP has anti-hyperglycemic, antioxidant, anti-inflammatory and antiapoptotic potentials, and exhibits synergistic effect with metformin.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Própole , Animais , Glicemia/metabolismo , Caspase 3/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Sinergismo Farmacológico , Inibidores de Glicosídeo Hidrolases/farmacologia , Insulina/sangue , Interleucina-1/metabolismo , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/fisiopatologia , Malásia , Masculino , Malondialdeído/metabolismo , NF-kappa B/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos Sprague-Dawley , Estreptozocina
17.
J Mol Endocrinol ; 60(3): 171-183, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29330151

RESUMO

Interleukin (IL)-22 has recently been suggested as an anti-inflammatory cytokine that could protect the islet cells from inflammation- and glucose-induced toxicity. We have previously shown that the tumor necrosis factor family member, LIGHT, can impair human islet function at least partly via pro-apoptotic effects. Herein, we aimed to investigate the protective role of IL-22 on human islets exposed to the combination of hyperglycemia and LIGHT. First, we found upregulation of LIGHT receptors (LTßR and HVEM) in engrafted human islets exposed to hyperglycemia (>11 mM) for 17 days post transplantation by using a double islet transplantation mouse model as well as in human islets cultured with high glucose (HG) (20 mM glucose) + LIGHT in vitro, and this latter effect was attenuated by IL-22. The effect of HG + LIGHT impairing glucose-stimulated insulin secretion was reversed by IL-22. The harmful effect of HG + LIGHT on human islet function seemed to involve enhanced endoplasmic reticulum stress evidenced by upregulation of p-IRE1α and BiP, elevated secretion of pro-inflammatory cytokines (IL-6, IL-8, IP-10 and MCP-1) and the pro-coagulant mediator tissue factor (TF) release and apoptosis in human islets, whereas all these effects were at least partly reversed by IL-22. Our findings suggest that IL-22 could counteract the harmful effects of LIGHT/hyperglycemia on human islet cells and potentially support the strong protective effect of IL-22 on impaired islet function and survival.


Assuntos
Apoptose/efeitos dos fármacos , Hiperglicemia/patologia , Interleucinas/farmacologia , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/toxicidade , Adulto , Idoso , Animais , Citocinas/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Receptores de Interleucina/metabolismo , Receptores do LH/genética , Receptores do LH/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Regulação para Cima/efeitos dos fármacos , Adulto Jovem , Interleucina 22
18.
J Inherit Metab Dis ; 41(1): 19-27, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29247329

RESUMO

Diabetes mellitus (DM) is not a single disease, but several pathophysiological conditions where synthesis, release, and/or action of insulin are disturbed. A progressive autoimmune/autoinflammatory destruction of islet cells is still considered the main pathophysiological event in the development of T1DM, but there is evidence that T1DM itself is a heterogeneous disease. More than 50 gene regions are closely associated with T1DM and a variety of epigenetic factors and metabolic patterns have been characterized, which may play a role in the development of T1DM. The pathogenesis and genetics of type 2 DM (T2DM) are distinct from T1DM. Genes associated with T2DM are distinct from those in T1DM. Characteristic metabolic patterns, different from those in T1DM were reported in T2DM, and some children with T2DM also express islet-antibodies. Huge progress has been made in the characterization of other specific types of DM, which had been considered very rare before. The molecular clarification of maturity-onset diabetes of the young (MODY) has greatly improved our understanding of the pathophysiology of DM. There are genetic overlaps between T2DM and monogenetic DM. Neonatal DM has been shown to be monogenetic in most cases, and genetic elucidation leads to more precise and individualized therapies. Cystic fibrosis related DM (CFRDM) should be considered a genuine part of cystic fibrosis, and not a complication, since pancreatic fibrosis does not sufficiently explain the pathophysiology of CFRDM. Disturbances of cystic fibrosis transmembrane conductance regulator (CFTR) as well as autoimmunity are involved in the pathogenesis of CFRDM.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Metabolismo Energético , Doenças do Recém-Nascido/fisiopatologia , Ilhotas Pancreáticas/fisiopatologia , Adolescente , Idade de Início , Autoanticorpos/imunologia , Autoimunidade , Glicemia/metabolismo , Criança , Pré-Escolar , Fibrose Cística/epidemiologia , Fibrose Cística/genética , Fibrose Cística/metabolismo , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/epidemiologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/genética , Metabolismo Energético/genética , Predisposição Genética para Doença , Humanos , Lactente , Recém-Nascido , Doenças do Recém-Nascido/sangue , Doenças do Recém-Nascido/epidemiologia , Doenças do Recém-Nascido/genética , Insulina/sangue , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Fatores de Risco
19.
Adv Exp Med Biol ; 1043: 385-399, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29224104

RESUMO

In rodent models of insulin-deficient diabetes, 17ß-estradiol (E2) protects pancreatic insulin-producing ß-cells against oxidative stress, amyloid polypeptide toxicity, gluco-lipotoxicity, and apoptosis. Three estrogen receptors (ERs)-ERα, ERß, and the G protein-coupled ER (GPER)-have been identified in rodent and human ß-cells. This chapter describes recent advances in our understanding of the role of ERs in islet ß-cell function, nutrient homeostasis, survival from pro-apoptotic stimuli, and proliferation. We discuss why and how ERs represent potential therapeutic targets for the maintenance of functional ß-cell mass.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Terapia de Reposição de Estrogênios , Estrogênios/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Apoptose , Glicemia/metabolismo , Proliferação de Células , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 1/terapia , Estradiol/efeitos adversos , Terapia de Reposição de Estrogênios/efeitos adversos , Estrogênios/efeitos adversos , Humanos , Hipoglicemiantes/uso terapêutico , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Ilhotas Pancreáticas/cirurgia , Transplante das Ilhotas Pancreáticas , Ligantes , Camundongos Endogâmicos NOD , Estado Nutricional , Receptores de Estrogênio/efeitos dos fármacos , Fatores de Risco , Transdução de Sinais
20.
Endocrinology ; 158(10): 3162-3173, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28938442

RESUMO

The metabolic syndrome is associated with an increase in the activation of the renin angiotensin system, whose inhibition reduces the incidence of new-onset diabetes. Importantly, angiotensin II (AngII), independently of its vasoconstrictor action, causes ß-cell inflammation and dysfunction, which may be an early step in the development of type 2 diabetes. The aim of this study was to determine how AngII causes ß-cell dysfunction. Islets of Langerhans were isolated from C57BL/6J mice that had been infused with AngII in the presence or absence of taurine-conjugated ursodeoxycholic acid (TUDCA) and effects on endoplasmic reticulum (ER) stress, inflammation, and ß-cell function determined. The mechanism of action of AngII was further investigated using isolated murine islets and clonal ß cells. We show that AngII triggers ER stress, an increase in the messenger RNA expression of proinflammatory cytokines, and promotes ß-cell dysfunction in murine islets of Langerhans both in vivo and ex vivo. These effects were significantly attenuated by TUDCA, an inhibitor of ER stress. We also show that AngII-induced ER stress is required for the increased expression of proinflammatory cytokines and is caused by reactive oxygen species and IP3 receptor activation. These data reveal that the induction of ER stress is critical for AngII-induced ß-cell dysfunction and indicates how therapies that promote ER homeostasis may be beneficial in the prevention of type 2 diabetes.


Assuntos
Angiotensina II/farmacologia , Estresse do Retículo Endoplasmático/fisiologia , Inflamação/fisiopatologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Animais , Linhagem Celular Tumoral , Citocinas/genética , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/prevenção & controle , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases/antagonistas & inibidores , Endorribonucleases/genética , Endorribonucleases/fisiologia , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glucose/farmacologia , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Insulinoma , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , RNA Interferente Pequeno , Espécies Reativas de Oxigênio/metabolismo , Sistema Renina-Angiotensina/fisiologia , Taurina/farmacologia , Ácido Ursodesoxicólico/farmacologia , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA