Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Discov Med ; 36(183): 655-665, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38665015

RESUMO

Incretin hormones, such as glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 and 2 (GLP-1, 2), belong to the group of gastrointestinal hormones. Their actions occur through interaction with GIP and GLP-1/2 receptors, which are present in various target tissues. Apart from their well-established roles in pancreatic function and insulin regulation, incretins elicit significant effects that extend beyond the pancreas. Specifically, these hormones stimulate osteoblast differentiation and inhibit osteoclast activity, thereby promoting bone anabolism. Moreover, they play a pivotal role in bone mineralization and overall bone quality and function, making them potentially therapeutic for managing bone health. Thus, this review provides a summary of the crucial involvement of incretins in bone metabolism, influencing both bone formation and resorption processes. While existing evidence is persuasive, further studies are necessary for a comprehensive understanding of the therapeutic potential of incretins in modifying bone health.


Assuntos
Remodelação Óssea , Polipeptídeo Inibidor Gástrico , Peptídeo 1 Semelhante ao Glucagon , Peptídeo 2 Semelhante ao Glucagon , Incretinas , Humanos , Remodelação Óssea/efeitos dos fármacos , Polipeptídeo Inibidor Gástrico/metabolismo , Incretinas/uso terapêutico , Incretinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Animais , Osso e Ossos/metabolismo , Osso e Ossos/efeitos dos fármacos , Pâncreas/metabolismo , Pâncreas/efeitos dos fármacos , Pâncreas/patologia
2.
J Clin Endocrinol Metab ; 108(5): 1084-1092, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-36458872

RESUMO

CONTEXT: A portion of ingested fats are retained in the intestine for many hours before they are mobilized and secreted in chylomicron (CM) particles. Factors such as glucagon-like peptide-2 (GLP-2) and glucose can mobilize these stored intestinal lipids and enhance CM secretion. We have recently demonstrated in rodents that GLP-2 acutely enhances CM secretion by mechanisms that do not involve the canonical CM synthetic assembly and secretory pathways. OBJECTIVE: To further investigate the mechanism of GLP-2's potent intestinal lipid mobilizing effect, we examined intracellular cytoplasmic lipid droplets (CLDs) in intestinal biopsies of humans administered GLP-2 or placebo. DESIGN, SETTING, PATIENTS, AND INTERVENTIONS: A single dose of placebo or GLP-2 was administered subcutaneously 5 hours after ingesting a high-fat bolus. In 1 subset of participants, plasma samples were collected to quantify lipid and lipoprotein concentrations for 3 hours after placebo or GLP-2. In another subset, a duodenal biopsy was obtained 1-hour after placebo or GLP-2 administration for transmission electron microscopy and proteomic analysis. RESULTS: GLP-2 significantly increased plasma triglycerides by 46% (P = 0.009), mainly in CM-sized particles by 133% (P = 0.003), without reducing duodenal CLD size or number. Several proteins of interest were identified that require further investigation to elucidate their potential role in GLP-2-mediated CM secretion. CONCLUSIONS: Unlike glucose that mobilizes enterocyte CLDs and enhances CM secretion, GLP-2 acutely increased plasma CMs without significant mobilization of CLDs, supporting our previous findings that GLP-2 does not act directly on enterocytes to enhance CM secretion and most likely mobilizes secreted CMs in the lamina propria and lymphatics.


Assuntos
Quilomícrons , Gotículas Lipídicas , Humanos , Quilomícrons/metabolismo , Triglicerídeos , Gotículas Lipídicas/metabolismo , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Proteômica , Glucose
3.
Endocrinology ; 163(4)2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35266539

RESUMO

Enterotoxigenic Escherichia coli (ETEC)-derived purified heat-stable enterotoxin b (STb) is responsible for secretory diarrhea in livestock and humans. STb disrupts intestinal fluid homeostasis, epithelial barrier function, and promotes cell death. Glucagon-like peptide-2 (GLP-2) is a potent intestinotrophic hormone secreted by enteroendocrine L cells. GLP-2 enhances crypt cell proliferation, epithelial barrier function, and inhibits enterocyte apoptosis. Whether STb can affect GLP-2 producing L cells remains to be elucidated. First, secreted-His-labeled STb from transformed E coli was collected and purified. When incubated with L-cell models (GLUTag, NCI-H716, and secretin tumor cell line [STC-1]), fluorescent immunocytochemistry revealed STb was internalized and was differentially localized in the cytoplasm and nucleus. Cell viability experiments with neutral red and resazurin revealed that STb was toxic in all but the GLUTag cells. STb stimulated 2-hour GLP-2 secretion in all cell models. Interestingly, GLUTag cells produced the highest amount of GLP-2 when treated with STb, demonstrating an inverse relationship in GLP-2 secretion and cell toxicity. To demonstrate a protective role for GLP-2, GLUTag-conditioned media (rich in GLP-2) blocked STb toxicity in STC-1 cells. Confirming a protective role of GLP-2, teduglutide was able to improve cell viability in cells treated with H2O2. In conclusion, STb interacts with the L cell, stimulates secretion, and may induce toxicity if GLP-2 is not produced at high levels. GLP-2 or receptor agonists have the ability to improve cell viability in response to toxins. These results suggest that GLP-2 secretion can play a protective role during STb intoxication. This work supports future investigation into the use of GLP-2 therapies in enterotoxigenic-related diseases.


Assuntos
Enterotoxinas , Peptídeo 2 Semelhante ao Glucagon , Animais , Células Enteroendócrinas/metabolismo , Enterotoxinas/metabolismo , Enterotoxinas/toxicidade , Escherichia coli/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Temperatura Alta , Humanos , Peróxido de Hidrogênio/metabolismo , Células L , Camundongos
4.
Cell Mol Gastroenterol Hepatol ; 13(6): 1829-1842, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35218981

RESUMO

BACKGROUND & AIMS: Leucine-rich repeat-containing G-protein-coupled receptor-5 (Lgr5)+/olfactomedin-4 (Olfm4)+ intestinal stem cells (ISCs) in the crypt base are crucial for homeostatic maintenance of the epithelium. The gut hormone, glucagon-like peptide-21-33 (GLP-2), stimulates intestinal proliferation and growth; however, the actions of GLP-2 on the Lgr5+ ISCs remain unclear. The aim of this study was to determine whether and how GLP-2 regulates Lgr5+ ISC cell-cycle dynamics and numbers. METHODS: Lgr5-Enhanced green-fluorescent protein - internal ribosome entry site - Cre recombinase - estrogen receptor T2 (eGFP-IRES-creERT2) mice were acutely administered human Glycine2 (Gly2)-GLP-2, or the GLP-2-receptor antagonist, GLP-23-33. Intestinal epithelial insulin-like growth factor-1-receptor knockout and control mice were treated chronically with human Gly2 (hGly2)-GLP-2. Cell-cycle parameters were determined by 5-Ethynyl-2'-deoxyuridine (EdU), bromodeoxyuridine, antibody #Ki67, and phospho-histone 3 labeling and cell-cycle gene expression. RESULTS: Acute hGly2-GLP-2 treatment increased the proportion of eGFP+EdU+/OLFM4+EdU+ cells by 11% to 22% (P < .05), without affecting other cell-cycle markers. hGly2-GLP-2 treatment also increased the ratio of eGFP+ cells in early to late S-phase by 97% (P < .001), and increased the proportion of eGFP+ cells entering S-phase by 218% (P < .001). hGly2-GLP-2 treatment induced jejunal expression of genes involved in cell-cycle regulation (P < .05), and increased expression of Mcm3 in the Lgr5-expressing cells by 122% (P < .05). Conversely, GLP-23-33 reduced the proportion of eGFP+EdU+ cells by 27% (P < .05), as well as the expression of jejunal cell-cycle genes (P < .05). Finally, chronic hGly2-GLP-2 treatment increased the number of OLFM4+ cells/crypt (P < .05), in an intestinal epithelial insulin-like growth factor-1-receptor-dependent manner. CONCLUSIONS: These findings expand the actions of GLP-2 to encompass acute stimulation of Lgr5+ ISC S-phase entry through the GLP-2R, and chronic induction of Lgr5+ ISC expansion through downstream intestinal insulin-like growth factor-1 signaling.


Assuntos
Peptídeo 2 Semelhante ao Glucagon , Fator de Crescimento Insulin-Like I , Animais , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Mucosa Intestinal , Intestinos , Camundongos , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo
5.
Peptides ; 148: 170683, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34748791

RESUMO

Glucose homeostasis is maintained by the glucoregulatory hormones, glucagon, insulin and somatostatin, secreted from the islets of Langerhans. Glucagon is the body's most important anti-hypoglycemic hormone, mobilizing glucose from glycogen stores in the liver in response to fasting, thus maintaining plasma glucose levels within healthy limits. Glucagon secretion is regulated by both circulating nutrients, hormones and neuronal inputs. Hormones that may regulate glucagon secretion include locally produced insulin and somatostatin, but also urocortin-3, amylin and pancreatic polypeptide, and from outside the pancreas glucagon-like peptide-1 and 2, peptide tyrosine tyrosine and oxyntomodulin, glucose-dependent insulinotropic polypeptide, neurotensin and ghrelin, as well as the hypothalamic hormones arginine-vasopressin and oxytocin, and calcitonin from the thyroid. Each of these hormones have distinct effects, ranging from regulating blood glucose, to regulating appetite, stomach emptying rate and intestinal motility, which makes them interesting targets for treating metabolic diseases. Awareness regarding the potential effects of the hormones on glucagon secretion is important since secretory abnormalities could manifest as hyperglycemia or even lethal hypoglycemia. Here, we review the effects of each individual hormone on glucagon secretion, their interplay, and how treatments aimed at modulating the plasma levels of these hormones may also influence glucagon secretion and glycemic control.


Assuntos
Glicemia/metabolismo , Glucagon/metabolismo , Pâncreas/metabolismo , Animais , Calcitonina/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Humanos , Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Neurotensina/metabolismo , Oxintomodulina/metabolismo , Ocitocina/metabolismo , Polipeptídeo Pancreático/metabolismo , Somatostatina/metabolismo , Urocortinas/metabolismo , Vasopressinas/metabolismo
6.
Front Endocrinol (Lausanne) ; 12: 697120, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34290670

RESUMO

Glucagon-like peptide-1 (GLP-1) shows robust protective effects on ß-cell survival and function and GLP-1 based therapies are successfully applied for type-2 diabetes (T2D) and obesity. Another cleavage product of pro-glucagon, Glucagon-like peptide-2 (GLP-2; both GLP-1 and GLP-2 are inactivated by DPP-4) has received little attention in its action inside pancreatic islets. In this study, we investigated GLP-2 production, GLP-2 receptor (GLP-2R) expression and the effect of GLP-2R activation in human islets. Isolated human islets from non-diabetic donors were exposed to diabetogenic conditions: high glucose, palmitate, cytokine mix (IL-1ß/IFN-γ) or Lipopolysaccharide (LPS) in the presence or absence of the DPP4-inhibitor linagliptin, the TLR4 inhibitor TAK-242, the GLP-2R agonist teduglutide and/or its antagonist GLP-2(3-33). Human islets under control conditions secreted active GLP-2 (full-length, non-cleaved by DPP4) into the culture media, which was increased by combined high glucose/palmitate, the cytokine mix and LPS and highly potentiated by linagliptin. Low but reproducible GLP-2R mRNA expression was found in all analyzed human islet isolations from 10 donors, which was reduced by pro-inflammatory stimuli: the cytokine mix and LPS. GLP-2R activation by teduglutide neither affected acute or glucose stimulated insulin secretion nor insulin content. Also, teduglutide had no effect on high glucose/palmitate- or LPS-induced dysfunction in cultured human islets but dampened LPS-induced macrophage-dependent IL1B and IL10 expression, while its antagonist GLP-2(3-33) abolished such reduction. In contrast, the expression of islet macrophage-independent cytokines IL6, IL8 and TNF was not affected by teduglutide. Medium conditioned by teduglutide-exposed human islets attenuated M1-like polarization of human monocyte-derived macrophages, evidenced by a lower mRNA expression of pro-inflammatory cytokines, compared to vehicle treated islets, and a reduced production of itaconate and succinate, marker metabolites of pro-inflammatory macrophages. Our results reveal intra-islet production of GLP-2 and GLP-2R expression in human islets. Despite no impact on ß-cell function, local GLP-2R activation reduced islet inflammation which might be mediated by a crosstalk between endocrine cells and macrophages.


Assuntos
Peptídeo 2 Semelhante ao Glucagon/metabolismo , Inflamação , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Macrófagos/fisiologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Células Cultivadas , Feminino , Receptor do Peptídeo Semelhante ao Glucagon 2/genética , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/fisiologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/efeitos dos fármacos , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Masculino , Pancreatite/imunologia , Pancreatite/metabolismo , Pancreatite/patologia
7.
PLoS One ; 15(10): e0239601, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33112876

RESUMO

APC mutations drive human colorectal cancer (CRC) development. A major contributing factor is colonic stem cell (SC) overpopulation. But, the mechanism has not been fully identified. A possible mechanism is the dysregulation of neuroendocrine cell (NEC) maturation by APC mutations because SCs and NECs both reside together in the colonic crypt SC niche where SCs mature into NECs. So, we hypothesized that sequential inactivation of APC alleles in human colonic crypts leads to progressively delayed maturation of SCs into NECs and overpopulation of SCs. Accordingly, we used quantitative immunohistochemical mapping to measure indices and proportions of SCs and NECs in human colon tissues (normal, adenomatous, malignant), which have different APC-zygosity states. In normal crypts, many cells staining for the colonic SC marker ALDH1 co-stained for chromogranin-A (CGA) and other NEC markers. In contrast, in APC-mutant tissues from familial adenomatous polyposis (FAP) patients, the proportion of ALDH+ SCs progressively increased while NECs markedly decreased. To explain how these cell populations change in FAP tissues, we used mathematical modelling to identify kinetic mechanisms. Computational analyses indicated that APC mutations lead to: 1) decreased maturation of ALDH+ SCs into progenitor NECs (not progenitor NECs into mature NECs); 2) diminished feedback signaling by mature NECs. Biological experiments using human CRC cell lines to test model predictions showed that mature GLP-2R+ and SSTR1+ NECs produce, via their signaling peptides, opposing effects on rates of NEC maturation via feedback regulation of progenitor NECs. However, decrease in this feedback signaling wouldn't explain the delayed maturation because both progenitor and mature NECs are depleted in CRCs. So the mechanism for delayed maturation must explain how APC mutation causes the ALDH+ SCs to remain immature. Given that ALDH is a key component of the retinoic acid (RA) signaling pathway, that other components of the RA pathway are selectively expressed in ALDH+ SCs, and that exogenous RA ligands can induce ALDH+ cancer SCs to mature into NECs, RA signaling must be attenuated in ALDH+ SCs in CRC. Thus, attenuation of RA signaling explains why ALDH+ SCs remain immature in APC mutant tissues. Since APC mutation causes increased WNT signaling in FAP and we found that sequential inactivation of APC in FAP patient tissues leads to progressively delayed maturation of colonic ALDH+ SCs, the hypothesis is developed that human CRC evolves due to an imbalance between WNT and RA signaling.


Assuntos
Transformação Celular Neoplásica/genética , Colo/citologia , Colo/metabolismo , Neoplasias Colorretais/genética , Genes APC , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Mutação , Somatostatina/metabolismo , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Família Aldeído Desidrogenase 1/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Cromogranina A/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Retroalimentação Fisiológica , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Células HCT116 , Células HT29 , Humanos , Camundongos , Modelos Genéticos , Células Neuroendócrinas/citologia , Células Neuroendócrinas/metabolismo , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Nicho de Células-Tronco , Tretinoína/metabolismo , Via de Sinalização Wnt
8.
Curr Osteoporos Rep ; 18(4): 371-377, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32504189

RESUMO

PURPOSE OF REVIEW: To provide an update on the acute effects of glucose, insulin, and incretins on markers of bone turnover in those with and without diabetes. RECENT FINDINGS: Bone resorption is suppressed acutely in response to glucose and insulin challenges in both healthy subjects and patients with diabetes. The suppression is stronger with oral glucose compared with intravenous delivery. Stronger responses with oral glucose may be related to incretin effects on insulin secretion or from a direct effect on bone turnover. Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-2 (GLP-2) infusion acutely suppresses bone resorption without much effect on bone formation. The bone turnover response to a metabolic challenge may be attenuated in type 2 diabetes, but this is an understudied area. A knowledge gap exists regarding bone turnover responses to a metabolic challenge in type 1 diabetes. The gut-pancreas-bone link is potentially an endocrine axis. This linkage is disrupted in diabetes, but the mechanism and progression of this disruption are not understood.


Assuntos
Remodelação Óssea/fisiologia , Reabsorção Óssea/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Incretinas/metabolismo , Insulina/metabolismo , Osteogênese/fisiologia , Estudos de Casos e Controles , Metabolismo Energético , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Humanos , Secreção de Insulina/fisiologia
9.
Acta Pharmacol Sin ; 41(11): 1446-1456, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32398684

RESUMO

Dipeptidyl peptidase 4 (DPP4), a ubiquitously expressed protease that cleaves off the N-terminal dipeptide from proline and alanine on the penultimate position, has important roles in many physiological processes. In the present study, experimental colitis was induced in mice receiving 3% dextran sulfate sodium (DSS) in drinking water. We found that mice with DSS-induced colitis had significantly increased intestinal DPP activity and decreased serum DPP activity, suggesting a probable correlation of DPP4 with experimental colitis. Then, we investigated whether sitagliptin, a specific DPP4 inhibitor could protect against DSS-induced colitis. We showed that oral administration of single dose of sitagliptin (30 mg/kg) on D7 remarkably inhibited DPP enzyme activity in both serum and intestine of DSS-induced colitic mice. Repeated administration of sitagliptin (10, 30 mg/kg, bid, from D0 to D8) significantly ameliorated DSS-induced colitis, including reduction of disease activity index (DAI) and body weight loss, improvement of histological score and colon length. Sitagliptin administration dose-dependently increased plasma concentrations of active form of GLP-1 and colonic expression of GLP-2R. Co-administration of GLP-2R antagonist GLP-23-33 (500 µg/kg, bid, sc) abolished the protective effects of sitagliptin in DSS-induced colitic mice. Moreover, sitagliptin administration significantly decreased the ratio of apoptotic cells and increased the ratio of proliferative cells in colon epithelium of DSS-induced colitic mice, and this effect was also blocked by GLP-23-33. Taken together, our results demonstrate that sitagliptin could attenuate DSS-induced experimental colitis and the effects can be attributed to the enhancement of GLP-2 action and the subsequent protective effects on intestinal barrier by inhibiting epithelial cells apoptosis and promoting their proliferation. These findings suggest sitagliptin as a novel therapeutic approach for the treatment of ulcerative colitis.


Assuntos
Colite/prevenção & controle , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Fosfato de Sitagliptina/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colite/induzido quimicamente , Colite/metabolismo , Citocinas/metabolismo , Sulfato de Dextrana , Dipeptidil Peptidase 4/metabolismo , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos BALB C , Junções Íntimas/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Dig Dis Sci ; 65(12): 3521-3537, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32072437

RESUMO

BACKGROUND: In short bowel syndrome, epithelial surface loss results in impaired nutrient absorption and may lead to intestinal insufficiency or intestinal failure. Nucleotide oligomerization domain 2 (Nod2) dysfunction predisposes to the development of intestinal failure after intestinal resection and is associated with intestinal barrier defects. Epithelial barrier function is crucial for intestinal absorption and for intestinal adaptation in the short bowel situation. AIMS: The aim of the study was to characterize the effects of the GLP-2 analogue Teduglutide in the small intestine in the presence and absence of Nod2 in a mouse model of short bowel syndrome. METHODS: Mice underwent 40% ICR and were thereafter treated with Teduglutide versus vehicle injections. Survival, body weight, stool water, and sodium content and plasma aldosterone concentrations were determined. Intestinal and kidney tissue was examined with light and fluorescence microscopy, Ussing chamber studies and quantitative PCR in wild type and transgenic mice. RESULTS: Teduglutide reduced intestinal failure incidence in Nod2 k.o. mice. In wt mice, Teduglutide attenuated intestinal insufficiency as indicated by reduced body weight loss and lower plasma aldosterone concentrations, lower stool water content, and lower stool sodium losses. Teduglutide treatment was associated with enhanced epithelial paracellular pore function and enhanced claudin-10 expression in tight junctions in the villus tips, where it colocalized with sodium-glucose cotransporter 1 (SGLT-1), which mediates Na-coupled glucose transport. CONCLUSIONS: In the SBS situation, Teduglutide not only maximizes small intestinal mucosal hypertrophy but also partially restores small intestinal epithelial function through an altered distribution of claudin-10, facilitating sodium recirculation for Na-coupled glucose transport and water absorption.


Assuntos
Proteína Adaptadora de Sinalização NOD2/metabolismo , Peptídeos/farmacologia , Síndrome do Intestino Curto/metabolismo , Animais , Modelos Animais de Doenças , Fármacos Gastrointestinais/farmacologia , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Junções Íntimas/metabolismo
11.
Dig Dis Sci ; 65(9): 2605-2618, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32006214

RESUMO

BACKGROUND: Circulating endotoxin (lipopolysaccharide, LPS) increases the gut paracellular permeability. We hypothesized that glucagon-like peptide-2 (GLP-2) acutely reduces LPS-related increased intestinal paracellular permeability by a mechanism unrelated to its intestinotrophic effect. METHODS: We assessed small intestinal paracellular permeability in vivo by measuring the appearance of intraduodenally perfused FITC-dextran 4000 (FD4) into the portal vein (PV) in rats 1-24 h after LPS treatment (5 mg/kg, ip). We also examined the effect of a stable GLP-2 analog teduglutide (TDG) on FD4 permeability. RESULTS: FD4 movement into the PV was increased 6 h, but not 1 or 3 h after LPS treatment, with increased PV GLP-2 levels and increased mRNA expressions of proinflammatory cytokines and proglucagon in the ileal mucosa. Co-treatment with a GLP-2 receptor antagonist enhanced PV FD4 concentrations. PV FD4 concentrations 24 h after LPS were higher than FD4 concentrations 6 h after LPS, reduced by exogenous GLP-2 treatment given 6 or 12 h after LPS treatment. FD4 uptake measured 6 h after LPS was reduced by TDG 3 or 6 h after LPS treatment. TDG-associated reduced FD4 uptake was reversed by the VPAC1 antagonist PG97-269 or L-NAME, not by EGF or IGF1 receptor inhibitors. CONCLUSIONS: Systemic LPS releases endogenous GLP-2, reducing LPS-related increased permeability. The therapeutic window of exogenous GLP-2 administration is at minimum within 6-12 h after LPS treatment. Exogenous GLP-2 treatment is of value in the prevention of increased paracellular permeability associated with endotoxemia.


Assuntos
Endotoxemia/prevenção & controle , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 2/agonistas , Absorção Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Dextranos/sangue , Modelos Animais de Doenças , Endotoxemia/sangue , Endotoxemia/induzido quimicamente , Fluoresceína-5-Isotiocianato/análogos & derivados , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Mediadores da Inflamação/metabolismo , Intestino Delgado/metabolismo , Lipopolissacarídeos , Masculino , Permeabilidade , Veia Porta , Ratos Sprague-Dawley , Fatores de Tempo
12.
BMC Cancer ; 19(1): 1016, 2019 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664952

RESUMO

BACKGROUND: Chemotherapy-induced alimentary mucositis (AM) is difficult to prevent and treatment is rarely effective. Recent study have been showed that glucagon-like peptide (GLP)-1 and GLP-2 has protective in chemotherapy-induced AM. While the DPP-4 enzyme degrades this GLP-1, the DPP-4 inhibitor blocks the degradation process and raises the concentration of GLP-1. This study aimed to assess the role of DPP-4 inhibitor, a well-known hypoglycemic agent, on chemotherapy-induced AM. METHODS: Twenty-four 6-week-old male C57BL/6 mice were divided into 4 groups: control, 5-fluorouracil (5-FU), DPP-4 inhibitor, and saline (DPP-4i), and DPP-4 inhibitor and 5-FU (DPP-4i + 5-FU). Mucositis was induced by intraperitoneal injection of 5-FU (400 mg/kg). DPP-4 inhibitor (50 mg/kg) was administered orally for four days starting the day before 5-FU administration. Post 72 h of 5-FU injection, mice were sacrificed and body weight change, diarrhea score, villus height, villus/crypt ratio, histologic characteristics including goblet cell count, and mRNA expression of inflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6, were assessed. RESULTS: Daily body weight change was not statistically significant between the 5-FU and the DPP-4i + 5-FU group (P = 0.571). Diarrhea score was significantly different between these two groups (P = 0.033). In the 5-FU group, the villus height was not maintained well, the epithelial lining was irregular, and inflammatory cell infiltration was observed. Goblet cell count in the DPP-4i + 5-FU group was significantly higher than in the 5-FU group (P = 0.007). However, in the DPP-4i + 5-FU group, the villus height, epithelial lining, and crypt structure were better maintained than in the 5-FU group. Compared with the control group, mRNA expression of TNF-α was significantly up-regulated in the 5-FU group. Moreover, mRNA expression of TNF-α in the DPP-4i + 5-FU group was down-regulated compared to the 5-FU group. However, IL-6 in the 5-FU group was significantly down-regulated compared to the control, there was no significant difference in expression of IL-6 between the 5-FU and DPP4i + 5-FU group. CONCLUSION: DPP-4 inhibitor can improve 5-FU induced AM and, therefore, has potential as an alternative treatment for chemotherapy-induced AM.


Assuntos
Antimetabólitos Antineoplásicos/efeitos adversos , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Fluoruracila/efeitos adversos , Mucosite/induzido quimicamente , Mucosite/tratamento farmacológico , Substâncias Protetoras/uso terapêutico , Administração Oral , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/uso terapêutico , Peso Corporal/efeitos dos fármacos , Diarreia/tratamento farmacológico , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/administração & dosagem , Modelos Animais de Doenças , Fluoruracila/administração & dosagem , Fluoruracila/uso terapêutico , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Células Caliciformes/efeitos dos fármacos , Injeções Intraperitoneais , Interleucina-6/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mucosite/patologia , Substâncias Protetoras/administração & dosagem , Fator de Necrose Tumoral alfa/genética
13.
Diabetes Obes Metab ; 21(11): 2535-2541, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31364232

RESUMO

AIM: To test the hypothesis that gut hormone glucagon-like peptide-2 (GLP-2) mobilizes intestinal triglyceride (TG) stores and stimulates chylomicron secretion by a nitric oxide (NO)-dependent mechanism in humans. METHODS: In a randomized, single-blind, cross-over study, 10 healthy male volunteers ingested a high-fat formula followed, 7 hours later, by one of three treatments: NO synthase inhibitor L-NG -monomethyl arginine acetate (L-NMMA) + GLP-2 analogue teduglutide, normal saline + teduglutide, or L-NMMA + placebo. TG in plasma and lipoprotein fractions were measured, along with measurement of blood flow in superior mesenteric and coeliac arteries using Doppler ultrasound in six participants. RESULTS: Teduglutide rapidly increased mesenteric blood flow and TG concentrations in plasma, in TG-rich lipoproteins, and most robustly in chylomicrons. L-NMMA significantly attenuated teduglutide-induced enhancement of mesenteric blood flow but not TG mobilization and chylomicron secretion. CONCLUSIONS: GLP-2 mobilization of TG stores and stimulation of chylomicron secretion from the small intestine appears to be independent of systemic NO in humans.


Assuntos
Peptídeo 2 Semelhante ao Glucagon/metabolismo , Mucosa Intestinal/metabolismo , Lipoproteínas/metabolismo , Óxido Nítrico/metabolismo , Triglicerídeos/metabolismo , Artéria Celíaca/diagnóstico por imagem , Quilomícrons/química , Quilomícrons/metabolismo , Humanos , Mucosa Intestinal/efeitos dos fármacos , Lipoproteínas/sangue , Masculino , Artéria Mesentérica Superior/diagnóstico por imagem , Pessoa de Meia-Idade , Peptídeos/farmacologia , Método Simples-Cego , Triglicerídeos/sangue , Ultrassonografia Doppler
14.
J Anim Physiol Anim Nutr (Berl) ; 103(5): 1338-1350, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31342562

RESUMO

The objective of this study was to test the hypothesis that aspartame supplementation in starter diet accelerates small intestinal cell cycle by stimulating secretion and expression of glucagon-like peptide -2 (GLP-2) in pre-weaned lambs using animal and cell culture experiments. In vivo, twelve 14-day-old lambs were selected and allocated randomly to two groups; one was treated with plain starter diet (Con, n = 6) and the other was treated with starter supplemented with 200 mg of aspartame/kg starter (APM, n = 6). Results showed that the lambs received APM treatment for 35 d had higher (p < .05) GLP-2 concentration in the plasma and greater jejunum weight/live body weight (BW) and jejunal crypt depth. Furthermore, APM treatment significantly upregulated (p < .05) the mRNA expression of cyclin D1 in duodenum; and cyclin A2, cyclin D1, cyclin-dependent kinases 6 (CDK6) in jejunum; and cyclin A2, cyclin D1, CDK4 in ileum. Moreover, APM treatment increased (p < .05) the mRNA expression of glucagon (GCG), insulin-like growth factor 1 (IGF-1) in the jejunum and ileum and mRNA expression of GLP-2 receptor (GLP-2R) in the jejunum. In vitro, when jejunal cells were treated with GLP-2 for 2 hr, the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) OD, IGF-1 concentration, and the mRNA expression of IGF-1, cyclin D1 and CDK6 were increased (p < .05). Furthermore, IGF-1 receptor (IGF-1R) inhibitor decreased (p < .05) the mRNA expression of IGF-1, cyclin A2, cyclin D1 and CDK6 in GLP-2 treatment jejunal cells. These results suggest that aspartame supplementation in starter accelerates small intestinal cell cycle that may, in part, be related to stimulate secretion and expression of GLP-2 in pre-weaning lambs. Furthermore, GLP-2 can indirectly promote the proliferation of jejunal cells mainly through the IGF-1 pathway. These findings provide new insights into nutritional interventions that promote the development of small intestines in young ruminants.


Assuntos
Aspartame/farmacologia , Células Epiteliais/efeitos dos fármacos , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Mucosa Intestinal/citologia , Intestino Delgado/efeitos dos fármacos , Ovinos/fisiologia , Ração Animal , Animais , Animais Lactentes , Aspartame/administração & dosagem , Células Cultivadas , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeo 2 Semelhante ao Glucagon/genética , Receptor do Peptídeo Semelhante ao Glucagon 2/genética , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Proglucagon/genética , Proglucagon/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo
15.
Yakugaku Zasshi ; 139(5): 783-791, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-31061348

RESUMO

This review focuses on the anti-dementia and antidepressant-like effects of peptides including glucagon-like peptide (GLP)-1, GLP-2, neuromedin U (NmU), and oxytocin, and the intranasal delivery of these peptides to the brain. Intracerebroventricularly administered GLP-1, NmU, and oxytocin improved impairment of learning and memory in mice treated with lipopolysaccharide or ß-amyloid protein. GLP-1 also improved impairment of learning and memory in juvenile diabetes model rats. On the other hand, GLP-2 exhibited antidepressant-like effects in mice during the forced-swim test, which were associated with 5-HT1A, α2, ß1, and D2 receptors. GLP-2 also exerted antidepressant-like effects in adrenocorticotropic hormone (ACTH)-treated mice through restoration of the hypothalamic-pituitary-adrenal-axis and neurogenesis in the subgranular zone of the dentate gyrus. Because intracerebroventricular administration is invasive and the peptides are unable to penetrate the blood-brain barrier, we introduced our new method of intranasal administration to deliver the peptides to the brain. We prepared a GLP-2 derivative containing cell-penetrating peptides (CPPs) and a penetration accelerating sequence (PAS). Intranasally administered PAS-CPPs-GLP-2 was distributed throughout the brain, and exhibited antidepressant-like effects in both naive and ACTH-treated mice. The derivatives of GLP-1, NmU, and oxytocin with the PAS and CPPs were also distributed throughout the brain after intranasal administration, and improved impairment of learning and memory. We confirmed that our peptide derivatives were effectively delivered into the brain by intranasal administration. As such, these derivatives may be useful for the clinical treatment of psychiatric and neurological diseases.


Assuntos
Administração Intranasal/métodos , Encéfalo/metabolismo , Demência/tratamento farmacológico , Depressão/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Peptídeos/administração & dosagem , Peptídeos/metabolismo , Animais , Peptídeo 1 Semelhante ao Glucagon/administração & dosagem , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/administração & dosagem , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Humanos , Camundongos , Neuropeptídeos/administração & dosagem , Neuropeptídeos/metabolismo , Ocitocina/administração & dosagem , Ocitocina/metabolismo , Ratos
16.
J Vis Exp ; (147)2019 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-31132057

RESUMO

Intestinal adaptation is the natural compensatory mechanism that occurs when the bowel is lost due to trauma. The adaptive responses, such as crypt cell proliferation and increased nutrient absorption, are critical in recovery, yet poorly understood. Understanding the molecular mechanism behind the adaptive responses is crucial to facilitate the identification of nutrients or drugs to enhance adaptation. Different approaches and models have been described throughout the literature, but a detailed descriptive way to essentially perform the procedures is needed to obtain reproducible data. Here, we describe a method to estimate important endpoints and proliferative markers of small intestinal injury and compensatory hyperproliferation using a model of chemotherapy-induced mucositis in mice. We demonstrate the detection of proliferating cells using a cell cycle specific marker, as well as using small intestinal weight, crypt depth, and villus height as endpoints. Some of the critical steps within the described method are the removal and weighing of the small intestine and the rather complex software system suggested for the measurement of this technique. These methods have the advantages that they are not time-consuming, and that they are cost-effective and easy to carry out and measure.


Assuntos
Adaptação Fisiológica , Antineoplásicos/efeitos adversos , Biomarcadores/metabolismo , Determinação de Ponto Final , Intestino Delgado/lesões , Mucosite/induzido quimicamente , Mucosite/patologia , Doença Aguda , Animais , Peso Corporal , Bromodesoxiuridina/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/patologia , Camundongos Endogâmicos C57BL
17.
Am J Physiol Gastrointest Liver Physiol ; 316(5): G668-G678, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30896970

RESUMO

Diabetes is a worldwide health problem. Roux-en-Y gastric bypass (RYGB) leads to rapid resolution of type 2 diabetes (T2D). Decreased hepatic insulin resistance is key, but underlying mechanisms are poorly understood. We hypothesized that changes in intestinal function and subsequent changes in portal venous milieu drive some of these postoperative benefits. We therefore aimed to evaluate postoperative changes in portal milieu. Two rat strains, healthy [Sprague-Dawley (SD)] and obese diabetic [Zucker diabetic fatty (ZDF)] rats, underwent RYGB or control surgery. After 4 wk, portal and systemic blood was sampled before and during an intestinal glucose bolus to investigate changes in intestinal glucose absorption (Gabsorp) and utilization (Gutil), and intestinal secretion of incretins and glucagon-like peptide-2 (GLP-2). Hepatic activity of dipeptidyl peptidase-4 (DPP4), which degrades incretins, was also measured. RYGB decreased Gabsorp in both rat strains. Gutil increased in SD rats and decreased in ZDF rats. In both strains, there was increased expression of intestinal hexokinase and gluconeogenesis enzymes. Systemic incretin and GLP-2 levels also increased after RYGB. This occurred without an increase in secretion. Hepatic DPP4 activity and expression were unchanged. RYGB perturbs multiple intestinal pathways, leading to decreased intestinal glucose absorption and increased incretin levels in both healthy and diabetic animals. In diabetic rats, intestinal glucose balance shifts toward glucose release. The portal vein as the gut-liver axis may integrate these intestinal changes to contribute to rapid changes in hepatic glucose and hormone handling. This fresh insight into the surgical physiology of RYGB raises the hope of less invasive alternatives. NEW & NOTEWORTHY Portal milieu after gastric bypass surgery is an underinvestigated area. Roux-en-Y gastric bypass perturbs multiple intestinal pathways, reducing intestinal glucose absorption and increasing incretin levels. In diabetic rats, the intestine becomes a net releaser of glucose, increasing portal glucose levels. The portal vein as the gut-liver axis may integrate these intestinal changes to contribute to changes in hepatic glucose handling. This fresh insight raises the hope of less invasive alternatives.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Derivação Gástrica , Glucose/metabolismo , Intestinos , Fígado , Sistema Porta/fisiologia , Animais , Diabetes Mellitus Experimental , Dipeptidil Peptidase 4/metabolismo , Células Enteroendócrinas/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Resistência à Insulina/fisiologia , Absorção Intestinal/fisiologia , Intestinos/irrigação sanguínea , Intestinos/cirurgia , Fígado/irrigação sanguínea , Fígado/metabolismo , Período Pós-Operatório , Ratos , Ratos Zucker
18.
J Clin Endocrinol Metab ; 104(7): 2463-2472, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30137354

RESUMO

CONTEXT: Several cases of cholelithiasis and cholecystitis have been reported in patients treated with glucagon-like peptide 1 (GLP-1) receptor agonists (GLP-1RAs) and GLP-2 receptor agonists (GLP-2RAs), respectively. Thus, the effects of GLP-1 and GLP-2 on gallbladder motility have been investigated. We have provided an overview of the mechanisms regulating gallbladder motility and highlight novel findings on the effects of bile acids and glucagon-like peptides on gallbladder motility. EVIDENCE ACQUISITION: The articles included in the present review were identified using electronic literature searches. The search results were narrowed to data reporting the effects of bile acids and GLPs on gallbladder motility. EVIDENCE SYNTHESIS: Bile acids negate the effect of postprandial cholecystokinin-mediated gallbladder contraction. Two bile acid receptors seem to be involved in this feedback mechanism, the transmembrane Takeda G protein-coupled receptor 5 (TGR5) and the nuclear farnesoid X receptor. Furthermore, activation of TGR5 in enteroendocrine L cells leads to release of GLP-1 and, possibly, GLP-2. Recent findings have pointed to the existence of a bile acid-TGR5-L cell-GLP-2 axis that serves to terminate meal-induced gallbladder contraction and thereby initiate gallbladder refilling. GLP-2 might play a dominant role in this axis by directly relaxing the gallbladder. Moreover, recent findings have suggested GLP-1RA treatment prolongs the refilling phase of the gallbladder. CONCLUSIONS: GLP-2 receptor activation in rodents acutely increases the volume of the gallbladder, which might explain the risk of gallbladder diseases associated with GLP-2RA treatment observed in humans. GLP-1RA-induced prolongation of human gallbladder refilling may explain the gallbladder events observed in GLP-1RA clinical trials.


Assuntos
Esvaziamento da Vesícula Biliar/efeitos dos fármacos , Vesícula Biliar/efeitos dos fármacos , Peptídeos Semelhantes ao Glucagon/efeitos adversos , Contração Muscular/efeitos dos fármacos , Ácidos e Sais Biliares/metabolismo , Colecistite/induzido quimicamente , Colecistite/fisiopatologia , Colecistocinina/metabolismo , Colelitíase/induzido quimicamente , Colelitíase/fisiopatologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Vesícula Biliar/fisiopatologia , Esvaziamento da Vesícula Biliar/fisiologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 2/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Humanos , Contração Muscular/fisiologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiopatologia , Obesidade/tratamento farmacológico , Período Pós-Prandial/fisiologia
19.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(3): 326-334, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30578967

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a continuum of disorders that can range from simple steatosis to non-alcoholic steatohepatitis (NASH). As a complex metabolic disorder, the pathophysiology of NAFLD is incompletely understood. Recently glucagon-like peptide (GLP)-1 and -2 signalling has been implicated in the pathogenesis of NAFLD. The role of these gut hormones in the hepatic abnormalities is complicated by lack of consensus on the presence of GLP-1 and GLP-2 receptors within the liver. Nevertheless, GLP-1 and GLP-2 receptor agonists have been associated with alterations in lipid metabolism and hepatic and systemic inflammation, pathological abnormalities characteristic of NAFLD. Treatment with GLP-1 analogues has been shown to reverse features of NAFLD including insulin resistance, and alterations in hepatic de novo lipogenesis and reactive oxygen species. In this review, we provide an overview of the role of GLP-1 and GLP-2 in lipid homeostasis and metabolic disease including NAFLD and NASH.


Assuntos
Peptídeos Semelhantes ao Glucagon/metabolismo , Metabolismo dos Lipídeos/fisiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 2/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Humanos , Resistência à Insulina , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/fisiologia , Lipogênese/efeitos dos fármacos , Lipogênese/fisiologia , Lipoproteínas/metabolismo , Liraglutida/farmacologia , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos
20.
PLoS One ; 13(6): e0198046, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29864142

RESUMO

OBJECTIVE: Mucositis is a side effect of chemotherapy seen in the digestive tract, with symptoms including pain, diarrhoea, inflammation and ulcerations. Our aim was to investigate whether endogenous glucagon-like peptide -1 and -2 (GLP-1 and GLP-2) are implicated in intestinal healing after chemotherapy-induced mucositis. DESIGN: We used a transgenic mouse model Tg(GCG.DTR)(Tg) expressing the human diphtheria toxin receptor in the proglucagon-producing cells. Injections with diphtheria toxin ablated the GLP-1 and GLP-2 producing L-cells in Tg mice with no effect in wild-type (WT) mice. Mice were injected with 5-fluorouracil or saline and received vehicle, exendin-4, teduglutide (gly2-GLP-2), or exendin-4/teduglutide in combination. The endpoints were body weight change, small intestinal weight, morphology, histological scoring of mucositis and myeloperoxidase levels. RESULTS: Ablation of L-cells led to impaired GLP-2 secretion; increased loss of body weight; lower small intestinal weight; lower crypt depth, villus height and mucosal area; and increased the mucositis severity score in mice given 5-fluorouracil. WT mice showed compensatory hyperproliferation as a sign of regeneration in the recovery phase. Co-treatment with exendin-4 and teduglutide rescued the body weight of the Tg mice and led to a hyperproliferation in the small intestine, whereas single treatment was less effective. CONCLUSION: The ablation of L-cells leads to severe mucositis and insufficient intestinal healing, shown by severe body weight loss and lack of compensatory hyperproliferation in the recovery phase. Co-treatment with exendin-4 and teduglutide could prevent this. Because both peptides were needed, we can conclude that both GLP-1 and GLP-2 are essential for intestinal healing in mice.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Mucosite/fisiopatologia , Regeneração , Animais , Sinergismo Farmacológico , Exenatida/farmacologia , Feminino , Fluoruracila/efeitos adversos , Peptídeo 1 Semelhante ao Glucagon/deficiência , Peptídeo 2 Semelhante ao Glucagon/deficiência , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mucosite/induzido quimicamente , Mucosite/metabolismo , Peptídeos/farmacologia , Regeneração/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA