Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 271
Filtrar
1.
J Clin Endocrinol Metab ; 109(8): 2003-2011, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38344778

RESUMO

CONTEXT: Distinguishing different types of diabetes is important in directing optimized treatment strategies and correlated epidemiological studies. OBJECTIVE: Through detailed analysis of hormone responses to mixed meal tolerance test (MMTT), we aimed to find representing characteristics of post-acute pancreatitis diabetes mellitus (PPDM-A) and post-chronic pancreatitis diabetes mellitus (PPDM-C). METHODS: Participants with PPDM-A, PPDM-C, type 1 diabetes, type 2 diabetes, and normal controls (NCs) underwent MMTT. Fasting and postprandial responses of serum glucose, C-peptide, insulin, glucagon, pancreatic polypeptide (PP), ghrelin, gastric inhibitory peptide (GIP), glucagon like peptide-1 (GLP-1), and peptide YY (PYY) were detected and compared among different groups. Focused analysis on calculated insulin sensitivity and secretion indices were performed to determine major causes of hyperglycemia in different conditions. RESULTS: Participants with PPDM-A were characterized by increased C-peptide, insulin, glucagon, and PP, but decreased ghrelin, GIP, and PYY compared with NCs. Patients with PPDM-C showed secretion insufficiency of C-peptide, insulin, ghrelin, and PYY, and higher postprandial responses of glucagon and PP than NCs. In particular, both fasting and postprandial levels of ghrelin in PPDM-C were significantly lower than other diabetes groups. PYY responses in patients with PPDM-A and PPDM-C were markedly reduced. Additionally, the insulin sensitivity of PPDM-A was decreased, and the insulin secretion for PPDM-C was decreased. CONCLUSION: Along with the continuum from acute to chronic pancreatitis, the pathological mechanism of PPDM changes from insulin resistance to insulin deficiency. Insufficient PYY secretion is a promising diagnostic marker for distinguishing PPDM from type 1 and type 2 diabetes. Absent ghrelin secretion to MMTT may help identify PPDM-C.


Assuntos
Grelina , Pancreatite , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Grelina/sangue , Diagnóstico Diferencial , Pancreatite/diagnóstico , Pancreatite/sangue , Pancreatite/etiologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Hormônios Gastrointestinais/sangue , Insulina/sangue , Peptídeo YY/sangue , Peptídeo C/sangue , Glucagon/sangue , Glicemia/análise , Glicemia/metabolismo , Período Pós-Prandial , Polipeptídeo Inibidor Gástrico/sangue , Peptídeo 1 Semelhante ao Glucagon/sangue , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/metabolismo , Estudos de Casos e Controles , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/sangue , Diabetes Mellitus/metabolismo , Hormônios Pancreáticos/sangue , Hormônios Pancreáticos/metabolismo , Resistência à Insulina
2.
Pancreas ; 53(3): e260-e267, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38345909

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease due to the lack of early detection. Because chronic pancreatitis (CP) patients are a high-risk group for pancreatic cancer, this study aimed to assess the differential miRNA profile in pancreatic tissue of patients with CP and pancreatic cancer. METHODS: MiRNAs were isolated from formalin-fixed paraffin-embedded pancreatic tissue of 22 PDAC patients, 18 CP patients, and 10 normal pancreatic tissues from autopsy (C) cases and processed for next-generation sequencing. Known and novel miRNAs were identified and analyzed for differential miRNA expression, target prediction, and pathway enrichment between groups. RESULTS: Among the miRNAs identified, 166 known and 17 novel miRNAs were found exclusively in PDAC tissues, while 106 known and 10 novel miRNAs were found specifically in CP tissues. The pathways targeted by PDAC-specific miRNAs and differentially expressed miRNAs between PDAC versus CP tissues and PDAC versus control tissues were the proteoglycans pathway, Hippo signaling pathway, adherens junction, and transforming growth factor-ß signaling pathway. CONCLUSIONS: This study resulted in a set of exclusive and differentially expressed miRNAs in PDAC and CP can be assessed for their diagnostic value. In addition, studying the role of miRNA-target gene interactions in carcinogenesis may open new therapeutic avenues.


Assuntos
Carcinoma Ductal Pancreático , MicroRNAs , Neoplasias Pancreáticas , Pancreatite Crônica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Pâncreas/patologia , Pancreatite Crônica/diagnóstico , Pancreatite Crônica/genética , Pancreatite Crônica/complicações , Hormônios Pancreáticos/metabolismo , Perfilação da Expressão Gênica
3.
Pancreas ; 52(1): e29-e36, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37378898

RESUMO

OBJECTIVES: The past studies clearly indicated that lumican was important in the context of pancreatic cancer (PC) onset and progression, but failed to clarify the underlying mechanistic basis for such activity. As such, we evaluated the functional importance of lumican in the context of pancreatic ductal adenocarcinoma (PDAC) to understand its mechanistic role in PC. METHODS: Lumican levels were evaluated in PDAC patient tissues via quantitative real-time polymerase chain reaction, Western blotting, and immunohistochemistry approaches. The role of lumican was additionally assessed via transfecting PDAC cell lines (BxPC-3, PANC-1) with lumican knockdown or overexpression constructs and treating PDAC cell lines with exogenous recombinant human lumican. RESULTS: Lumican expression levels were significantly higher in pancreatic tumor tissues relative to healthy paracancerous tissues. Lumican knockdown in BxPC-3 and PANC-1 enhanced their proliferation and migration, but reduced cellular apoptosis. Alternatively, lumican overexpression and exogenous lumican exposure failed to alter the proliferative activity of these cells. Further, lumican knockdown in BxPC-3 and PANC-1 cells results in marked P53 and P21 dysregulation. CONCLUSIONS: Lumican may suppress PDAC tumor growth by regulating P53 and P21, and the function of lumican sugar chains in the context of PC is worth studying in future studies.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Lumicana/genética , Lumicana/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias Pancreáticas/patologia , Pâncreas/patologia , Carcinoma Ductal Pancreático/patologia , Hormônios Pancreáticos/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas
4.
Peptides ; 158: 170893, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36244579

RESUMO

Chromogranin A (CgA) is a 439 amino acid protein secreted by neuroendocrine cells. Proteolytic processing of CgA results in the production of different bioactive peptides. These peptides have been associated with inflammatory bowel disease, diabetes, and cancer. One of the chromogranin A-derived peptides is ∼52 amino acid long Pancreastatin (PST: human (h)CgA250-301, murine (m)CgA263-314). PST is a glycogenolytic peptide that inhibits glucose-induced insulin secretion from pancreatic islet ß-cells. In addition to this metabolic role, evidence is emerging that PST also has inflammatory properties. This review will discuss the immunomodulatory properties of PST and its possible mechanisms of action and regulation. Moreover, this review will discuss the potential translation to humans and how PST may be an interesting therapeutic target for treating inflammatory diseases.


Assuntos
Cromograninas , Hormônios Pancreáticos , Humanos , Animais , Camundongos , Cromogranina A/farmacologia , Hormônios Pancreáticos/metabolismo , Cromograninas/metabolismo , Peptídeos , Aminoácidos
5.
Int J Mol Sci ; 23(19)2022 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-36233022

RESUMO

Oncogenic K-ras is often activated in pancreatic ductal adenocarcinoma (PDAC) due to frequent mutation (>90%), which drives multiple cellular processes, including alterations in lipid metabolism associated with a malignant phenotype. However, the role and mechanism of the altered lipid metabolism in K-ras-driven cancer remains poorly understood. In this study, using human pancreatic epithelial cells harboring inducible K-rasG12D (HPNE/K-rasG12D) and pancreatic cancer cell lines, we found that the expression of phospholipase A2 group IIA (PLA2G2A) was upregulated by oncogenic K-ras. The elevated expression of PLA2G2A was also observed in pancreatic cancer tissues and was correlated with poor survival of PDAC patients. Abrogation of PLA2G2A by siRNA or by pharmacological inhibition using tanshinone I significantly increased lipid peroxidation, reduced fatty acid synthase (FASN) expression, and impaired mitochondrial function manifested by a decrease in mitochondrial transmembrane potential and a reduction in ATP production, leading to the inhibition of cancer cell proliferation. Our study suggests that high expression of PLA2G2A induced by oncogenic K-ras promotes cancer cell survival, likely by reducing lipid peroxidation through its ability to facilitate the removal of polyunsaturated fatty acids from lipid membranes by enhancing the de novo fatty acid synthesis and energy metabolism to support cancer cell proliferation. As such, PLA2G2A might function as a downstream mediator of K-ras and could be a potential therapeutic target.


Assuntos
Carcinoma Ductal Pancreático , Fosfolipases A2 do Grupo II , Neoplasias Pancreáticas , Trifosfato de Adenosina/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Metabolismo Energético , Ácidos Graxos , Fosfolipases A2 do Grupo II/metabolismo , Humanos , Lipídeos , Mutação , Hormônios Pancreáticos/metabolismo , Neoplasias Pancreáticas/patologia , RNA Interferente Pequeno/metabolismo , Neoplasias Pancreáticas
6.
Nat Cancer ; 3(8): 945-960, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35982178

RESUMO

Cancer-associated fibroblasts (CAFs) are one of the most prominent and active components in the pancreatic tumor microenvironment. Our data show that CAFs are critical for survival from pancreatic ductal adenocarcinoma (PDAC) on glutamine deprivation. Specifically, we uncovered a role for nucleosides, which are secreted by CAFs through autophagy in a nuclear fragile X mental retardation-interacting protein 1 (NUFIP1)-dependent manner, increased glucose utilization and promoted growth of PDAC. Moreover, we demonstrate that CAF-derived nucleosides induced glucose consumption under glutamine-deprived conditions and displayed a dependence on MYC. Using an orthotopic mouse model of PDAC, we found that inhibiting nucleoside secretion by targeting NUFIP1 in the stroma reduced tumor weight. This finding highlights a previously unappreciated metabolic network within pancreatic tumors in which diverse nutrients are used to promote growth in an austere tumor microenvironment.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Autofagia , Fibroblastos Associados a Câncer/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proliferação de Células , Glucose/farmacologia , Glutamina/metabolismo , Camundongos , Proteínas Nucleares/metabolismo , Nucleosídeos/metabolismo , Hormônios Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
7.
Int J Mol Sci ; 23(10)2022 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-35628269

RESUMO

Elevated levels of Mucin-16 (MUC16) in conjunction with a high expression of truncated O-glycans is implicated in playing crucial roles in the malignancy of pancreatic ductal adenocarcinoma (PDAC). However, the mechanisms by which such aberrant glycoforms present on MUC16 itself promote an increased disease burden in PDAC are yet to be elucidated. This study demonstrates that the CRISPR/Cas9-mediated genetic deletion of MUC16 in PDAC cells decreases tumor cell migration. We found that MUC16 enhances tumor malignancy by activating the integrin-linked kinase and focal adhesion kinase (ILK/FAK)-signaling axis. These findings are especially noteworthy in truncated O-glycan (Tn and STn antigen)-expressing PDAC cells. Activation of these oncogenic-signaling pathways resulted in part from interactions between MUC16 and integrin complexes (α4ß1), which showed a stronger association with aberrant glycoforms of MUC16. Using a monoclonal antibody to functionally hinder MUC16 significantly reduced the migratory cascades in our model. Together, these findings suggest that truncated O-glycan containing MUC16 exacerbates malignancy in PDAC by activating FAK signaling through specific interactions with α4 and ß1 integrin complexes on cancer cell membranes. Targeting these aberrant glycoforms of MUC16 can aid in the development of a novel platform to study and treat metastatic pancreatic cancer.


Assuntos
Antígeno Ca-125 , Carcinoma Ductal Pancreático , Quinase 1 de Adesão Focal , Integrina alfa4beta1 , Proteínas de Membrana , Neoplasias Pancreáticas , Antígeno Ca-125/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/metabolismo , Humanos , Integrina alfa4beta1/metabolismo , Proteínas de Membrana/metabolismo , Hormônios Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Polissacarídeos/metabolismo
8.
Pancreas ; 51(10): 1411-1426, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37099787

RESUMO

OBJECTIVES: We evaluated the potential differentiation ability of bone mesenchymal stromal cells (BMSCs) into pancreatic lineage cells on a rat acellular pancreatic bioscaffold (APB) and the effect of differentiated BMSCs in vivo. METHODS: The BMSCs were dynamically or statically cultured with or without growth factor in both culture systems. We assessed the cytological behavior and differentiation. We also evaluated the pancreatic fibrosis and pathological scores. RESULTS: The proliferation rates of BMSCs were significantly higher in the APB groups. The APB induced BMSCs to express mRNA markers at higher levels. All tested pancreatic functional proteins were also expressed at higher levels in the APB group. The secretion of metabolic enzymes was higher in the APB system. The ultrastructure of BMSCs in the APB group further revealed the morphological characteristics of pancreatic-like cells. For the in vivo study, the pancreatic fibrosis and pathological scores were significantly lower in the differentiated BMSCs group. In addition, in both the in vitro and the in vivo study, growth factor significantly improved proliferation, differentiation, and pancreatic cell therapy. CONCLUSIONS: The APB can promote BMSC differentiation toward pancreatic lineage and pancreatic-like phenotypes, giving it the potential for use in pancreatic cell therapies and tissue engineering.


Assuntos
Medula Óssea , Células-Tronco Mesenquimais , Ratos , Animais , Células Cultivadas , Diferenciação Celular , Hormônios Pancreáticos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fibrose , Células Estromais
9.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36613557

RESUMO

A significant lack of donor organs restricts the opportunity to obtain tissue-specific scaffolds for tissue-engineering technologies. One of the acceptable solutions is the development of decellularization protocols for a human donor pancreas unsuitable for transplantation. A protocol of obtaining a biocompatible tissue-specific scaffold from decellularized fragments with pronounced human pancreas lipomatosis signs with preserved basic fibrillary proteins of a pancreatic tissue extracellular matrix was developed. The scaffold supports the adhesion and proliferation of human adipose derived stem cell (hADSCs) and prolongs the viability and insulin-producing function of pancreatic islets. Experiments conducted allow for the reliance on the prospects of using the donor pancreas unsuitable for transplantation in the technologies of tissue engineering and regenerative medicine, including the development of a tissue equivalent of a pancreas.


Assuntos
Ilhotas Pancreáticas , Pâncreas , Humanos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Matriz Extracelular/metabolismo , Hormônios Pancreáticos/metabolismo
10.
Peptides ; 132: 170366, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32634450

RESUMO

Amylin is a peptide hormone that is mainly known to be produced by pancreatic ß-cells in response to a meal but amylin is also produced by brain cells in discrete brain areas albeit in a lesser amount. Amylin receptor (AMY) is composed of the calcitonin core-receptor (CTR) and one of the 3 receptor activity modifying protein (RAMP), thus forming AMY1-3; RAMP enhances amylin binding properties to the CTR. However, amylin receptor agonist such as salmon calcitonin is able to bind CTR alone. Peripheral amylin's main binding site is located in the area postrema (AP) which then propagate the signal to the nucleus of the solitary tract and lateral parabrachial nucleus (LPBN) and it is then transmitted to the forebrain areas such as central amygdala and bed nucleus of the stria terminalis. Amylin's activation of these different brain areas mediates eating and other metabolic pathways controlling energy expenditure and glucose homeostasis. Peripheral amylin can also bind in the arcuate nucleus of the hypothalamus where it acts independently of the AP to activate POMC and NPY neurons. Amylin activation of NPY neurons has been shown to be transmitted to LPBN neurons to act on eating while amylin POMC signaling affects energy expenditure and locomotor activity. While a large amount of experiments have already been conducted, future studies will have to further investigate how amylin is taken up by forebrain areas and deepen our understanding of amylin action on peripheral metabolism.


Assuntos
Depressores do Apetite/metabolismo , Encéfalo/metabolismo , Ingestão de Alimentos/fisiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Animais , Humanos , Hormônios Pancreáticos/metabolismo , Transdução de Sinais
11.
Pancreatology ; 20(5): 813-821, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32522508

RESUMO

BACKGROUND/OBJECTIVES: Endocrine insufficiency following severe acute pancreatitis (SAP) leads to diabetes of the exocrine pancreas, (type 3c diabetes mellitus), however it is not known how this metabolic phenotype differs from that of type 2 diabetes, or how the two subtypes can be differentiated. We sought to determine the prevalence of diabetes following SAP, and to analyse the behaviour of glucose and pancreatic hormones across a 2-h oral glucose tolerance test (OGTT). METHODS: Twenty-six patients following SAP (mean (range) duration of first SAP episode to study time of 119.3 (14.8-208.9) months) along with 26 matched controls underwent an OGTT with measurement of glucose, insulin, c-peptide, glucagon and pancreatic polypeptide (PP) at fasting/15/90/120min. Beta-cell area was estimated using the 15min c-peptide/glucose ratio, and insulin resistance (IR) using homeostasis model assessment (HOMA) and oral glucose insulin sensitivity (OGIS) models. RESULTS: The prevalence of diabetes/prediabetes was 54% following SAP (38.5% newly-diagnosed compared to 19.2% newly-diagnosed controls). Estimated beta-cell area and IR did not differ between groups. AUC c-peptide was lower in SAP versus controls. AUC insulin and AUC c-peptide were lower in SAP patients with diabetes versus controls with diabetes; between-group differences were observed at the 90 and 120 min time-points only. Half of new diabetes cases in SAP patients were only identified at the 120min timepoint. CONCLUSIONS: Diabetes and pre-diabetes occur frequently following SAP and are difficult to distinguish from type 2 diabetes in controls but are characterised by reduced insulin and c-peptide at later stages of an OGTT. Consistent with this observation, most new post SAP diabetes cases were diagnosed by 2-h glucose levels only.


Assuntos
Diabetes Mellitus/epidemiologia , Diabetes Mellitus/etiologia , Doenças Metabólicas/epidemiologia , Doenças Metabólicas/etiologia , Pancreatite/complicações , Pancreatite/epidemiologia , Doença Aguda , Adulto , Idoso , Glicemia/metabolismo , Peptídeo C/sangue , Estudos de Casos e Controles , Feminino , Seguimentos , Teste de Tolerância a Glucose , Hemoglobinas Glicadas/análise , Humanos , Resistência à Insulina , Células Secretoras de Insulina/patologia , Masculino , Pessoa de Meia-Idade , Hormônios Pancreáticos/metabolismo , Estado Pré-Diabético/epidemiologia , Estado Pré-Diabético/etiologia , Prevalência
12.
Pancreas ; 49(6): 806-811, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32541637

RESUMO

OBJECTIVE: When total pancreatectomy with islet autotransplantation (TPIAT) is performed for chronic pancreatitis, the pancreas and most of the duodenum are removed, with Roux-en-Y reconstruction of the gastrointestinal tract. Enteroendocrine cells in the intestines and pancreas secrete hormones coordinating digestion and motility, but anatomic reconstruction alters transit of nutrients to these cells. We hypothesized that TPIAT leads to changes in enteroendocrine hormones. METHODS: Glucagon-like peptide 1 (GLP-1), peptide YY (PYY), and pancreatic polypeptide (PP) were measured from mixed-meal tolerance tests of 34 clinical trial participants before and 18 months after TPIAT. Area under the curve of GLP-1 and PYY-stimulated responses were calculated by trapezoidal method, and the PP response was measured as the stimulated max minus baseline (ΔPP). RESULTS: Area under the curve of GLP-1 and PYY increased significantly after TPIAT (GLP-1 average +553.1 pg/mL per minute, P = 0.004; PYY average +4647.9 pg/mL per minute, P = 0.02). ΔPP trended toward lower after TPIAT (average, -52.2 pg/mL, P = 0.06). CONCLUSIONS: In this novel study of enteroendocrine hormones in TPIAT patients, stimulated levels of GLP-1 and PYY were significantly higher after versus before TPIAT. ΔPP was lower after TPIAT, but not significantly. These hormone changes have potential clinical implications that warrant further research.


Assuntos
Células Enteroendócrinas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Pancreatectomia/métodos , Pancreatite Crônica/cirurgia , Adulto , Feminino , Hormônios Gastrointestinais/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Hormônios Pancreáticos/metabolismo , Polipeptídeo Pancreático/metabolismo , Peptídeo YY/metabolismo , Transplante Autólogo
13.
J Clin Endocrinol Metab ; 105(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32222768

RESUMO

CONTEXT: Impaired glucose homeostasis is a common finding in pheochromocytoma (PHEO), especially with adrenergic phenotype. The possible contribution of incretin dysfunction to dysglycemia in PHEO patients has not been studied. OBJECTIVE: To compare changes in pancreatic endocrine function and gut hormones' production during a liquid meal test before and 1 year after adrenalectomy. METHODS: In a prospective study, we included 18 patients with PHEO (13 females) with adrenergic biochemical phenotype. A liquid meal test with predefined isocaloric enteral nutrition was performed to evaluate dynamic changes in pancreatic hormones and incretins. RESULTS: During the meal test, insulin levels were significantly lower before adrenalectomy only in the early phase of insulin secretion, but changes in area under the curve (AUC) did not reach statistical significance (AUC = 0.07). Plasma glucagon (AUC < 0.01) and pancreatic polypeptide levels (AUC < 0.01) were suppressed in comparison with the postoperative state. Impaired response to the meal was found preoperatively for glucagon-like peptide-1 (GLP-1; AUC P < 0.05), but not glucose-dependent insulinotropic polypepide (GIP; AUC P = 0.21). No significant changes in insulin resistance indices were found, except for the homeostatic model assessment-beta index, an indicator of the function of islet ß cells, which negatively correlated with plasma metanephrine (R = -0.66, P < 0.01). CONCLUSIONS: Our study shows suppression of pancreatic α and ß cell function and impaired GLP-1 secretion during a dynamic meal test in patients with PHEO, which is improved after its surgical treatment. These data demonstrate a novel and potentially significant interconnection between excessive catecholamine production and the secretion of glucoregulatory hormones.


Assuntos
Neoplasias das Glândulas Suprarrenais/complicações , Adrenérgicos/efeitos adversos , Biomarcadores/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Intolerância à Glucose/etiologia , Feocromocitoma/complicações , Adulto , Idoso , Glicemia/análise , Feminino , Seguimentos , Hormônios Gastrointestinais/metabolismo , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Humanos , Incretinas/efeitos adversos , Masculino , Refeições , Pessoa de Meia-Idade , Hormônios Pancreáticos/metabolismo , Fenótipo , Prognóstico , Estudos Prospectivos
14.
Compr Physiol ; 10(2): 577-595, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32163198

RESUMO

GLP-1 was described as an incretin over 30 years ago. GLP-1 is encoded by the preproglucagon gene (Gcg), which is expressed in the intestine, the pancreas, and the central nervous system. GLP-1 activates GLP-1 receptors (GLP-1r) on the ß-cell to induce insulin secretion in a glucose-dependent manner. GLP-1 also inhibits α-cell secretion of glucagon. As few, if any, GLP-1r are expressed on α-cells, indirect regulation, via ß- or δ-cell products has been thought to be the primary mechanism by which GLP-1 inhibits glucagon secretion. However, recent work suggests that there is sufficient expression of GLP-1r on α-cells for direct regulation as well. Although the predominant source of circulating GLP-1 is the intestine, the α-cell becomes a source of GLP-1 when the islet is metabolically stressed. Recent work suggests the possibility that this source of GLP-1 is also be important in regulating nutrient-induced insulin secretion in a paracrine fashion. More work is also accumulating regarding the role of glucagon, another Gcg-derived protein produced by the α-cell, in stimulating insulin secretion by acting on GLP-1r. Altogether, these data clearly demonstrate the important role of Gcg-derived peptides in regulating insulin secretion. Because of GLP-1's important role in glucose homeostasis, it has been implicated in the success of bariatric surgery and has been successfully targeted for the treatment of type 2 diabetes mellitus. © 2020 American Physiological Society. Compr Physiol 10:577-595, 2020.


Assuntos
Diabetes Mellitus/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células Secretoras de Glucagon/metabolismo , Hormônios Pancreáticos/metabolismo , Animais , Homeostase/fisiologia , Humanos
15.
Front Endocrinol (Lausanne) ; 11: 608248, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424773

RESUMO

Obesity and obesity-related diseases are major public health concerns that have been exponentially growing in the last decades. Bariatric surgery is an effective long-term treatment to achieve weight loss and obesity comorbidity remission. Post-bariatric hypoglycemia (PBH) is a late complication of bariatric surgery most commonly reported after Roux-en-Y gastric bypass (RYGB). PBH is the end result of postprandial hyperinsulinemia but additional endocrine mechanisms involved are still under debate. Our aim was to characterize entero-pancreatic hormone dynamics associated with postprandial hypoglycemia after RYGB. Individuals previously submitted to RYGB (N=23) in a single tertiary hospital presenting PBH symptoms (Sym, n=14) and asymptomatic weight-matched controls (Asy, n=9) were enrolled. Participants underwent a mixed-meal tolerance test (MMTT) to assess glucose, total amino acids (total AA), insulin, C-peptide, glucagon, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and neurotensin (NT). We found that hypoglycemia during the MMTT was equally frequent in Sym and Asy groups (p=1.000). Re-grouped according to glucose nadir during the MMTT (Hypo n=11 vs NoHypo n=12; nadir <3.05 mmol/l vs ≥3.05 mmol/l), subjects presented no differences in anthropometric (BMI: p=0.527) or metabolic features (HbA1c: p=0.358), yet distinct meal-elicited hormone dynamics were identified. Postprandial glucose excursion and peak glucose levels were similar (p>0.05), despite distinct late glycemic outcomes (t=60 min and t=90 min: p<0.01), with overall greater glycemic variability in Hypo group (minimum-to-maximum glucose ratio: p<0.001). Hypo group meal-triggered hormone profile was characterized by lower early glucagon (t=15 min: p<0.01) and higher insulin (t=30 min: p<0.05, t=45 min: p<0.001), C-peptide (t=30 min: p<0.01, t=45 min: p<0.001, t=60 min: p<0.05), and GLP-1 (t=45 min: p<0.05) levels. Hyperinsulinemia was an independent risk factor for hypoglycemia (p<0.05). After adjusting for hyperinsulinemia, early glucagon correlated with glycemic nadir (p<0.01), and prevented postprandial hypoglycemia (p<0.05). A higher insulin to glucagon balance in Hypo was observed (p<0.05). No differences were observed in total AA, GIP or NT excursions (p>0.05). In sum, after RYGB, postprandial hyperinsulinemia is key in triggering PBH, but a parallel and earlier rise in endogenous glucagon might sustain the inter-individual variability in glycemic outcome beyond the effect of hyperinsulinism, advocating a potential pivotal role for glucagon in preventing hyperinsulinemic hypoglycemia.


Assuntos
Cirurgia Bariátrica , Glucagon/fisiologia , Hipoglicemia/etiologia , Hipoglicemia/prevenção & controle , Complicações Pós-Operatórias/prevenção & controle , Adulto , Aminoácidos/sangue , Aminoácidos/metabolismo , Glicemia/análise , Índice de Massa Corporal , Estudos de Coortes , Feminino , Glucagon/sangue , Teste de Tolerância a Glucose , Humanos , Hiperinsulinismo/metabolismo , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Hormônios Pancreáticos/metabolismo , Período Pós-Prandial
16.
Surg Obes Relat Dis ; 14(10): 1521-1529, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30449509

RESUMO

BACKGROUND: Changes in gastrointestinal and pancreatic hormones may play a role in promoting long-term weight reduction and improved glucose metabolism after sleeve gastrectomy and Roux-en-Y gastric bypass. However, few studies have examined the metabolic and endocrine effects of these procedures in Mainland China. OBJECTIVES: To compare the effects of laparoscopic sleeve gastrectomy (LSG) and laparoscopic Roux-en-Y gastric bypass (LRYGB) on gastrointestinal and pancreatic peptide hormones. SETTING: University hospital, China. METHODS: A nonrandomized prospective study was conducted in Chinese obese patients undergoing LSG or LRYGB. Of 20 patients in this study, 10 underwent LSG, and 10 underwent LRYGB. Fasting plasma levels of insulin, glucagon, ghrelin, gastric inhibitory peptide, peptide YY, glucagon-like peptide (GLP)-1, and GLP-2 were measured preoperatively and at 1, 3, 6, and 12 months after surgery. This trial was registered at www.clinicaltrials.gov (NCT02963662). RESULTS: During the first year after both operations, mean body mass index and fasting insulin levels steadily decreased at all intervals. Fasting plasma glucose levels significantly decreased at 1 month after surgery, then remained stable in both groups. Glucagon levels significantly decreased at 1, 3, and 6 months after surgery in both groups, but returned to baseline at 12 months. Fasting GLP-1 and peptide YY significantly increased in both groups, but more so after LRYGB. However, GLP-2 did not change in either group. Ghrelin levels significantly decreased after LSG, but not after LRYGB. Gastric inhibitory peptide levels decreased after LRYGB but not after LSG. CONCLUSIONS: LSG and LRYGB resulted in significant and distinct changes in multiple gastrointestinal and pancreatic peptide hormones that are important regulators of obesity and metabolic health.


Assuntos
Cirurgia Bariátrica/métodos , Gastrectomia/métodos , Hormônios Gastrointestinais/metabolismo , Laparoscopia/métodos , Hormônios Pancreáticos/metabolismo , Adulto , Biomarcadores/metabolismo , Glicemia/metabolismo , Índice de Massa Corporal , Jejum/sangue , Feminino , Derivação Gástrica/métodos , Glucagon/metabolismo , Humanos , Insulina/metabolismo , Masculino , Obesidade Mórbida/sangue , Obesidade Mórbida/cirurgia , Estudos Prospectivos , Redução de Peso/fisiologia
17.
Biochim Biophys Acta ; 1860(9): 2008-16, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27180174

RESUMO

BACKGROUND: Transplantation of islets of Langerhans is regarded as a promising therapy for type 1 diabetes. A large number of ß-cells are required for the treatment of human type 1 diabetes. Pluripotent stem cells, such as embryonic stem cells and induced pluripotent stem cells, have been considered as new sources for cell replacement therapy. METHODS: Cell aggregates were prepared from human iPS cells using agarose microwell plates and differentiated into pancreatic endocrine cells by changing the culture media with different additives. RESULTS: After 20days of culture, approximately 30% of cells in aggregates were positive for C-peptide. After another 14days in culture, the cells gained an ability to alter C-peptide release in response to changes in the glucose concentration. CONCLUSIONS: Uniform aggregates of human iPSCs were easily prepared on agarose microwell plates and efficiently differentiated into the pancreatic endocrine lineage. Thus, aggregate culture is a suitable method for preparing islet-like aggregates from human iPSCs. GENERAL SIGNIFICANCE: Our results indicate that the microwell plate is suitable for scaling up the preparation of pancreatic endocrine cells from human iPS cells in a robotic system.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Hormônios Pancreáticos/metabolismo , Peptídeo C/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Endócrinas/citologia , Células Endócrinas/metabolismo , Glucose , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
19.
J Am Coll Surg ; 222(4): 534-42, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26827125

RESUMO

BACKGROUND: Neuroendocrine tumors (NETs) of the duodenum are rare, heterogeneous, and often indolent neoplasms. We hypothesized that elevated pancreastatin levels are an indicator of a poor prognosis in well-differentiated duodenal NETs. STUDY DESIGN: Data from patients diagnosed with a primary duodenal NET were analyzed. Patients that underwent esophogogastroduodenoscopy, endoscopic ultrasound, or exploratory surgery to localize their neoplasm and whose tumors were confirmed histologically were included. RESULTS: Eighty-four patients were diagnosed with duodenal NETs from January 1991 to January 2014. Seventy-five percent and 21% of patients had their tumor localized by esophogogastroduodenoscopy and endoscopic ultrasound, respectively. The remaining 4% were localized during exploratory surgery. The 5-year Kaplan-Meier survival rate for the entire cohort (N = 84) was 80%. Survival sorted by normal vs abnormal pancreastatin level was statistically significant (p < 0.0001). Five-year survival rates were 94% and 37% for normal and abnormal pancreastatin, respectively. In contrast, survival sorted by normal vs abnormal plasma chromogranin A level was not statistically significant (p = 0.24). CONCLUSIONS: Patients with primary duodenal NETs have high 5-year survival rates. Serial monitoring of plasma pancreastatin levels can identify patients who have a poor prognosis.


Assuntos
Cromogranina A/metabolismo , Neoplasias Duodenais/metabolismo , Neoplasias Duodenais/mortalidade , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/mortalidade , Hormônios Pancreáticos/metabolismo , Idoso , Neoplasias Duodenais/cirurgia , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/cirurgia , Valor Preditivo dos Testes , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida
20.
J Clin Endocrinol Metab ; 101(2): 445-52, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26672633

RESUMO

CONTEXT: As a group, neuroendocrine tumors (NETs) secrete many different peptide hormones, yet heretofore each NET patient is typically thought to produce at most one hormone that causes a distinct hormonal syndrome. A minority of patients have multiple hormones at diagnosis and may also develop secondary hormone secretion at a later stage. OBJECTIVES: The objectives of the study were to determine the frequency and to describe the impact of multiple and secondary hormone secretion in sporadic gasteroenteropancreatic NET patients. DESIGN, SETTING, AND PARTICIPANTS: This was a retrospective analysis of patients (n = 972) with gasteroenteropancreatic NET treated at Uppsala University Hospital, Uppsala, Sweden. Patients with the secretion of multiple hormones at diagnosis and/or those developing secondary hormone secretion during the disease course were identified and studied in further detail. RESULTS: In pancreatic NETs (PNETs), a total of 19 of 323 patients (6%) had secretion of multiple hormones at diagnosis, and 14 of 323 (4%) had secondary changes during the disease course. These phenomena occurred exclusively in patients with an advanced disease stage, and secondary hormones were detected in a close time span with progressive disease. Patients with secondary insulin hypersecretion had increased morbidity as well as reduced survival (P < .002). In contrast, multiple and secondary hormone secretion was rarely seen in NETs of the small intestine with 0 and 1 of 603 cases, respectively. CONCLUSION: Diversity of PNET hormone secretion either at diagnosis or during the disease course occurred in a minority of patients (9.3%). These phenomena had a major impact on patient outcome both through increased morbidity and mortality. Our results support that patients with metastatic PNETs should be monitored for clinical symptoms of secondary hormone secretion during the disease course.


Assuntos
Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/secundário , Hormônios Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/secundário , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Progressão da Doença , Feminino , Neoplasias Gastrointestinais/patologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Intestino Delgado/metabolismo , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/mortalidade , Neoplasias Pancreáticas/mortalidade , Estudos Retrospectivos , Análise de Sobrevida , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA