Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Cell Physiol ; 236(9): 6507-6519, 2021 09.
Article in English | MEDLINE | ID: mdl-33559155

ABSTRACT

The intestinal mucosa is in continuous contact with milliard of microorganisms, thus intestinal epithelial barrier is a critical component in the arsenal of defense mechanisms required to prevent infection and inflammation. Mucin 2 (MUC2), which is produced by the goblet cells, forms the skeleton of the intestinal mucus and protects the intestinal tract from self-digestion and numerous microorganisms. Dedicator of cytokinesis 4 (DOCK4) is a member of the DOCK-B subfamily of the DOCK family of guanine nucleotide exchange factors. It is reported that DOCK4 plays a critical role in the repair of the barrier function of the intestinal epithelium after chemical damage. In this study, the role of DOCK4 in the goblet cell differentiation and MUC2 production is explored. Disordered intestinal epithelium and shortage of goblet cells were observed in DOCK4 gene knockout mice. Furthermore, DOCK4 deletion contributed to the low expression of MUC2 and the goblet cell differentiation/maturation factors including growth factor independent 1 (Gfi1) and SAM pointed domain epithelial-specific transcription factor (Spdef) in mouse ileums and colons. Overexpression of DOCK4 caused a marked increase in Gfi1, Spdef, and MUC2, while siRNA knockdown of endogenous DOCK4 significantly decreased Gfi1, Spdef, and MUC2 in HT-29 cells. In addition, MUC2, DOCK4, and the goblet cell differentiation/maturation factors mRNA levels were decreased in colorectal cancer samples compared with normal colons. A significant positive correlation was found between MUC2 and DOCK4. In conclusion, DOCK4 may serve as a critical regulator of goblet cell differentiation and MUC2 production in the intestine.


Subject(s)
Cell Differentiation , GTPase-Activating Proteins/metabolism , Goblet Cells/cytology , Goblet Cells/metabolism , Mucin-2/biosynthesis , Animals , Cell Differentiation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , GTPase-Activating Proteins/genetics , Gene Expression Regulation , HT29 Cells , Humans , Intestinal Mucosa/pathology , Mice, Knockout , Models, Biological
2.
J Am Chem Soc ; 143(38): 15635-15643, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34541841

ABSTRACT

Understanding the fundamental insights of oxygen activation and reaction at metal-oxide interfaces is of significant importance yet remains a major challenge due to the difficulty in in situ characterization of active oxygen species. Herein, the activation and reaction of molecular oxygen during CO oxidation at platinum-ceria interfaces has been in situ explored using surface-enhanced Raman spectroscopy (SERS) via a borrowing strategy, and different active oxygen species and their evolution during CO oxidation at platinum-ceria interfaces have been directly observed. In situ Raman spectroscopic evidence with isotopic exchange experiments demonstrate that oxygen is efficiently dissociated to chemisorbed O on Pt and lattice Ce-O species simultaneously at interfacial Ce3+ defect sites under CO oxidation, leading to a much higher activity at platinum-ceria interfaces compared to that at Pt alone. Further in situ time-resolved SERS studies and density functional theory simulations reveal a more efficient molecular pathway through the reaction between adsorbed CO and chemisorbed Pt-O species transferred from the interfaces. This work deepens the fundamental understandings on oxygen activation and CO oxidation at metal-oxide interfaces and offers a sensitive technique for the in situ characterization of oxygen species under working conditions.

3.
Angew Chem Int Ed Engl ; 60(37): 20535-20542, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34288301

ABSTRACT

The Co-based electrocatalyst is among the most promising candidates for electrochemical oxidation of 5-hydroxymethylfurfural (HMF). However, the intrinsic active sites and detailed mechanism of this catalyst remains unclear. We combine experimental evidence and a theoretical study to show that electrogenerated Co3+ and Co4+ species act as chemical oxidants but with distinct roles in selective HMF oxidation. It is found that Co3+ is only capable of oxidizing formyl group to produce carboxylate while Co4+ is required for the initial oxidation of hydroxyl group with significantly faster kinetics. As a result, the product distribution shows explicit dependence on the Co oxidation states and selective production of 5-hydroxymethyl-2-furancarboxylic acid (HMFCA) and 2,5-furandicarboxylic acid (FDCA) are achieved by tuning the applied potential. This work offers essential mechanistic insight on Co-catalyzed organic oxidation reactions and might guide the design of more efficient electrocatalysts.

4.
Sheng Li Xue Bao ; 72(3): 371-381, 2020 Jun 25.
Article in Zh | MEDLINE | ID: mdl-32572434

ABSTRACT

Caloric restriction (CR) is explored to limit the caloric intake without malnutrition. CR can affect the levels of various metabolites in organism, such as lipids, free fatty acids, ketones, bile acids and amino acids, etc, and is thought being able to extend the lifespan, postpone and reduce the incidence of age-related disorders (e.g., type 2 diabetes, cancer and cardiovascular diseases). These effects are mainly attributed to the role of CR in energy metabolism. The mechanism of CR on energy metabolism is closely related to biological clock, hormonal production, gastrointestinal flora and inflammation. Here we briefly review the effects and mechanism of CR on energy metabolism.


Subject(s)
Caloric Restriction , Diabetes Mellitus, Type 2 , Energy Metabolism , Humans , Longevity
5.
Cell Physiol Biochem ; 51(2): 664-680, 2018.
Article in English | MEDLINE | ID: mdl-30463068

ABSTRACT

BACKGROUND/AIMS: Roux-en-Y Gastric Bypass, RYGB, is the most effective strategy to control body weight in morbid obesity. RYGB leads to rapid improvement of glycemic status and weight loss, which are largely attributed to the alteration of gastrointestinal hormones including ghrelin. The current study examined potential mechanisms of altered ghrelin synthesis after RYGB. METHODS: Gastric mammalian target of rapamycin (mTOR) signaling, ghrelin synthesis and secretion were determined in lean or obese male mice with or without RYGB operation, as well as in obese patients pre- and post-RYGB surgery. Ghrelin expression and mTOR signaling were investigated by western blotting and immunohistochemistry. Ghrelin mRNA levels were detected by real-time PCR. Plasma ghrelin was measured by enzyme immunoassay. RESULTS: mTOR activity in the gastric fundus was significantly lower than in the forestomachs. Both of them were decreased after 24h fasting. A significant negative correlation was found between gastric levels of phospho-S6 (phospho-S6 ribosomal protein) and proghrelin during changes of energy status. mTOR activity was activated, whereas ghrelin expression was inhibited by Roux-en-Y Gastric Bypass in both rodents and human beings. Increment of ghrelin synthesis and decline of mTOR signaling induced by rapamycin were significantly reversed by RYGB in both lean and obese mice. Administration of Ad-S6K1 (adenovirus-mediated p70 ribosomal protein subunit 6 kinase 1) from tail vein suppressed the expression of ghrelin in RYGB-operated mice relative to control animals. CONCLUSION: mTOR is therefore a gastric fuel sensor whose activity is linked to the regulation of ghrelin after Roux-en-Y Gastric Bypass.


Subject(s)
Ghrelin/blood , Mechanistic Target of Rapamycin Complex 1/metabolism , Obesity, Morbid/pathology , Adult , Animals , Body Weight , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diet, High-Fat , Energy Metabolism , Gastric Bypass , Gastric Fundus/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Male , Mice, Inbred C57BL , Middle Aged , Obesity, Morbid/complications , Obesity, Morbid/surgery , Signal Transduction/drug effects , Sirolimus/pharmacology
6.
Proc Natl Acad Sci U S A ; 111(36): 13163-8, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25157160

ABSTRACT

Although ghrelin has been demonstrated to stimulate energy intake and storage through a central mechanism, its effect on hepatic lipid metabolism remains largely uncharacterized. Ghrelin receptor antagonism or gene deletion significantly decreased obesity-associated hepatic steatosis by suppression of de novo lipogenesis, whereas exogenous ghrelin stimulated lipogenesis, leading to hepatic lipid accumulation in mice. The effects of ghrelin were mediated by direct activation of its receptor on hepatocytes. Cultured hepatocytes responded to ghrelin with increased lipid content and expression of lipogenesis-related genes. Ghrelin increased phosphorylation of S6, the downstream target of mammalian target of rapamycin (mTOR) signaling in cultured hepatocytes, whereas ghrelin receptor antagonism reduced hepatic phosphorylation of S6 in db/db mice. Inhibition of mTOR signaling by rapamycin markedly attenuated ghrelin-induced up-regulation of lipogenesis in hepatocytes, whereas activation of hepatic mTOR signaling by deletion of TSC1 increased hepatic lipogenesis. By interacting with peroxisome proliferator-activated receptor-γ (PPARγ), mTOR mediates the ghrelin-induced up-regulation of lipogenesis in hepatocytes. The stimulatory effect of ghrelin on hepatic lipogenesis was significantly attenuated by PPARγ antagonism in cultured hepatocytes and in PPARγ gene-deficient mice. Our study indicates that ghrelin activates its receptor on hepatocytes to promote lipogenesis via a mechanism involving the mTOR-PPARγ signaling pathway.


Subject(s)
Ghrelin/pharmacology , Lipogenesis/drug effects , Liver/metabolism , PPAR gamma/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Deletion , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/drug effects , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/drug effects , Receptors, Ghrelin/metabolism
7.
Diabetologia ; 58(8): 1887-97, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26037201

ABSTRACT

AIMS/HYPOTHESIS: Glucagon-like peptide (GLP-1), an intestinal incretin produced in L cells through proglucagon processing, is released in response to meal intake. The intracellular mechanism by which L cells sense the organism energy level to coordinate the production of GLP-1 remains unclear. Mechanistic target of rapamycin (mTOR) is an intracellular fuel sensor critical for energy homeostasis. In this study, we investigated whether intestinal mTOR regulates GLP-1 production in L cells. METHODS: The effects of mTOR on GLP-1 production were examined in lean- or high-fat diet (HFD) induced diabetic C57/BL6, db/db, Neurog3-Tsc1(-/-) mice, and STC-1 cells. GLP-1 expression was investigated by real-time PCR and western blotting. Plasma GLP-1 and insulin were detected by enzyme immunoassay and radioimmunoassay, respectively. RESULTS: Fasting downregulated mTOR activity, which was associated with a decrement of intestinal proglucagon and circulating GLP-1. Upon re-feeding, these alterations returned to the levels of fed animals. In HFD induced diabetic mice, ileal mTOR signalling, proglucagon and circulating GLP-1 were significantly decreased. Inhibition of mTOR signalling by rapamycin decreased levels of intestinal and plasma GLP-1 in both normal and diabetic mice. Activation of the intestinal mTOR signalling by L-leucine or Tsc1 gene deletion increased levels of intestinal proglucagon and plasma GLP-1. Overexpression of mTOR stimulated proglucagon promoter activity and GLP-1 production, whereas inhibition of mTOR activity by overexpression of tuberous sclerosis 1 (TSC1) or TSC2 decreased proglucagon promoter activity and GLP-1 production in STC-1 cells. CONCLUSIONS/INTERPRETATION: mTOR may link energy supply with the production of GLP-1 in L cells.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Intestinal Mucosa/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat , Down-Regulation , Enteroendocrine Cells/drug effects , Fasting/metabolism , Intestines/drug effects , Leucine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proglucagon/metabolism , Sirolimus/pharmacology
8.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167185, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653360

ABSTRACT

OBJECTIVE: Glucagon is a critical hormone regulating glucose metabolism. It stimulates the liver to release glucose under low blood sugar conditions, thereby maintaining blood glucose stability. Excessive glucagon secretion and hyperglycemia is observed in individuals with diabetes. Precise modulation of glucagon is significant to maintain glucose homeostasis. Piezo1 is a mechanosensitive ion channel capable of converting extracellular mechanical forces into intracellular signals, thus regulating hormonal synthesis and secretion. This study aims to investigate the role of Piezo1 in regulating glucagon production in α cells. METHODS: The effects of Piezo1 on glucagon production were examined in normal- or high-fat diet fed α cell-specific Piezo1 knockout mice (Gcg-Piezo1-/-), and the murine pancreatic α cell line αTC1-6. Expression of Proglucagon was investigated by real-time PCR and western blotting. Plasma glucagon and insulin were detected by enzyme immunoassay. RESULTS: Under both normal- and high-fat diet conditions, Gcg-Piezo1-/- mice exhibited increased pancreatic α cell proportion, hyperglucagonemia, impaired glucose tolerance, and activated pancreatic mTORC1 signaling. Activation of Piezo1 by its agonist Yoda1 or overexpression of Piezo1 led to decreased glucagon synthesis and suppressed mTOR signaling pathway in αTC1-6 cells. Additionally, the levels of glucagon in the medium were also reduced. Conversely, knockdown of Piezo1 produced opposite effects. CONCLUSION: Our study uncovers the regulatory role of the Piezo1 ion channel in α cells. Piezo1 influences glucagon production by affecting mTOR signaling pathway.


Subject(s)
Diet, High-Fat , Glucagon-Secreting Cells , Glucagon , Ion Channels , Mice, Knockout , Animals , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Mice , Ion Channels/metabolism , Ion Channels/genetics , Diet, High-Fat/adverse effects , Male , Signal Transduction , Insulin/metabolism , Cell Line , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanotransduction, Cellular , Mice, Inbred C57BL , Proglucagon/metabolism , Proglucagon/genetics , Pyrazines , Thiadiazoles
9.
Abdom Radiol (NY) ; 49(2): 611-624, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38051358

ABSTRACT

PURPOSE: Microvascular invasion (MVI) is a common complication of hepatocellular carcinoma (HCC) surgery, which is an important predictor of reduced surgical prognosis. This study aimed to develop a fully automated diagnostic model to predict pre-surgical MVI based on four-phase dynamic CT images. METHODS: A total of 140 patients with HCC from two centers were retrospectively included (training set, n = 98; testing set, n = 42). All CT phases were aligned to the portal venous phase, and were then used to train a deep-learning model for liver tumor segmentation. Radiomics features were extracted from the tumor areas of original CT phases and pairwise subtraction images, as well as peritumoral features. Lastly, linear discriminant analysis (LDA) models were trained based on clinical features, radiomics features, and hybrid features, respectively. Models were evaluated by area under curve (AUC), accuracy, sensitivity, specificity, positive and negative predictive values (PPV and NPV). RESULTS: Overall, 86 and 54 patients with MVI- (age, 55.92 ± 9.62 years; 68 men) and MVI+ (age, 53.59 ± 11.47 years; 43 men) were included. Average dice coefficients of liver tumor segmentation were 0.89 and 0.82 in training and testing sets, respectively. The model based on radiomics (AUC = 0.865, 95% CI: 0.725-0.951) showed slightly better performance than that based on clinical features (AUC = 0.841, 95% CI: 0.696-0.936). The classification model based on hybrid features achieved better performance in both training (AUC = 0.955, 95% CI: 0.893-0.987) and testing sets (AUC = 0.913, 95% CI: 0.785-0.978), compared with models based on clinical and radiomics features (p-value < 0.05). Moreover, the hybrid model also provided the best accuracy (0.857), sensitivity (0.875), and NPV (0.917). CONCLUSION: The classification model based on multimodal intra- and peri-tumoral radiomics features can well predict HCC patients with MVI.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Male , Humans , Middle Aged , Aged , Adult , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/surgery , Radiomics , Retrospective Studies , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery , Tomography, X-Ray Computed
10.
Nat Metab ; 6(3): 458-472, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38467889

ABSTRACT

Ghrelin, produced mainly by gastric X/A-like cells, triggers a hunger signal to the central nervous system to stimulate appetite. It remains unclear whether X/A-like cells sense gastric distention and thus regulate ghrelin production. Here we show that PIEZO1 expression in X/A-like cells decreases in patients with obesity when compared to controls, whereas it increases after sleeve gastrectomy. Male and female mice with specific loss of Piezo1 in X/A-like cells exhibit hyperghrelinaemia and hyperphagia and are more susceptible to overweight. These phenotypes are associated with impairment of the gastric CaMKKII/CaMKIV-mTOR signalling pathway. Activation of PIEZO1 by Yoda1 or gastric bead implantation inhibits ghrelin production, decreases energy intake and induces weight loss in mice. Inhibition of ghrelin production by Piezo1 through the CaMKKII/CaMKIV-mTOR pathway can be recapitulated in a ghrelin-producing cell line mHypoE-42. Our study reveals a mechanical regulation of ghrelin production and appetite by PIEZO1 of X/A-like cells, which suggests a promising target for anti-obesity therapy.


Subject(s)
Ghrelin , TOR Serine-Threonine Kinases , Humans , Male , Female , Mice , Animals , Ghrelin/metabolism , TOR Serine-Threonine Kinases/metabolism , Obesity/metabolism , Appetite/physiology , Eating , Ion Channels/genetics
11.
Acta Pharm Sin B ; 13(1): 192-203, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36815030

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM), a fatal heart disease characterized by fibroadipocytic replacement of cardiac myocytes, accounts for 20% of sudden cardiac death and lacks effective treatment. It is often caused by mutations in desmosome proteins, with Desmoglein-2 (DSG2) mutations as a common etiology. However, the mechanism underlying the accumulation of fibrofatty in ACM remains unknown, which impedes the development of curative treatment. Here we investigated the fat accumulation and the underlying mechanism in a mouse model of ACM induced by cardiac-specific knockout of Dsg2 (CS-Dsg2 -/-). Heart failure and cardiac lipid accumulation were observed in CS-Dsg2 -/- mice. We demonstrated that these phenotypes were caused by decline of fatty acid (FA) ß-oxidation resulted from impaired mammalian target of rapamycin (mTOR) signaling. Rapamycin worsened while overexpression of mTOR and 4EBP1 rescued the FA ß-oxidation pathway in CS-Dsg2 -/- mice. Reactivation of PPARα by fenofibrate or AAV9-Pparα significantly alleviated the lipid accumulation and restored cardiac function. Our results suggest that impaired mTOR-4EBP1-PPARα-dependent FA ß-oxidation contributes to myocardial lipid accumulation in ACM and PPARα may be a potential target for curative treatment of ACM.

12.
Small Methods ; : e2301504, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38148311

ABSTRACT

Developing efficient oxygen evolution reaction (OER) electrocatalysts can greatly advance the commercialization of proton exchange membrane (PEM) water electrolysis. However, the unclear and disputed reaction mechanism and structure-activity relationship of OER pose significant obstacles. Herein, the active site and intermediate for OER on AuIr nanoalloys are simultaneously identified and correlated with the activity, through the integration of in situ shell-isolated nanoparticle-enhanced Raman spectroscopy and X-ray absorption spectroscopy. The AuIr nanoalloys display excellent OER performance with an overpotential of only 246 mV to achieve 10 mA cm-2 and long-term stability under strong acidic conditions. Direct spectroscopic evidence demonstrates that * OO adsorbed on IrOx sites is the key intermediate for OER, and it is generated through the O-O coupling of adsorbed oxygen species directly from water, providing clear support for the adsorbate evolution mechanism. Moreover, the Raman information of the * OO intermediate can serve as a universal "in situ descriptor" that can be obtained both experimentally and theoretically to accelerate the catalyst design. It unveils that weakening the interactions of * OO on the catalysts and facilitating its desorption would boost the OER performance. This work deepens the mechanistic understandings on OER and provides insightful guidance for the design of more efficient OER catalysts.

13.
Cell Physiol Biochem ; 29(3-4): 493-500, 2012.
Article in English | MEDLINE | ID: mdl-22508056

ABSTRACT

BACKGROUND: Nesfatin-1, an 82 amino acid peptide derived from the prohormone nucleobindin-2 (NUCB2), is a novel satiety hormone acting through a leptin-independent mechanism in the hypothalamus. The mechanisms by which production of nesfatin-1/NUCB2 is regulated remain unknown. METHODS: Nesfatin-1/NUCB2 mRNA and immunoreactivity were examined in gastric tissue and Min-6 cells by RT-PCR and immunofluorescent staining or Western blotting. RESULTS: Nesfatin-1/NUCB2 is co-localized with pS6K1, the downstream target of mammalian target of rapamycin (mTOR), in gastric X/A like cells. A parallel relationship between gastric mTOR signaling and nesfatin-1/NUCB2 was observed during changes in energy status. Both mTOR activity and gastric nesfatin-1/NUCB2 were down-regulated by fasting, and returned to basal levels with re-feeding. In high fat diet induced obese mice, gastric mTOR signaling and nesfatin-1/NUCB2 were increased. Inhibition of the gastric mTOR signaling by rapamycin attenuated the expression of gastric nesfatin-1/NUCB2 mRNA and protein in both lean and obese mice. Attenuation of mTOR activity by rapamycin or over-expression of TSC1 or TSC2 reduced the expression of nesfatin-1/NUCB2 in Min-6 cells, suggesting a direct effect of mTOR signaling. CONCLUSION: Gastric mTOR is a gastric energy sensor whose activity is linked to the regulation of gastric nesfatin-1/NUCB2.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Gastric Mucosa/metabolism , Nerve Tissue Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Calcium-Binding Proteins/genetics , Cell Line , DNA-Binding Proteins/genetics , Diet, High-Fat/adverse effects , Endocrine Cells/metabolism , Enzyme Activation , Fasting/metabolism , Gastric Mucosa/pathology , Gene Expression Regulation , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Nerve Tissue Proteins/genetics , Nucleobindins , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
14.
Genes (Basel) ; 14(1)2022 12 20.
Article in English | MEDLINE | ID: mdl-36672747

ABSTRACT

BACKGROUND: Both caloric restriction (CR) and Roux-en-Y gastric bypass (RYGB) are practical interventions for type 2 diabetes mellitus (T2DM), while the molecular mechanisms of CR and RYGB regarding glycemic control are still poorly understood. Here, we explore the effects and underlying mechanisms of CR and RYGB on ß-cell area and function. METHODS: Average islet size was measured by histological analysis. The pancreatic lipid content was detected by using a commercial lipid assay kit. The expression levels of lipogenic transcription factors and enzymes in mouse pancreas were determined by quantitative PCR, Western blot, and immunofluorescence. RESULTS: CR decreased the mean size of islets and pancreatic insulin production in both regular diet-fed and high-fat diet-fed mice. Increased ß-cell apoptosis was detected in the calorie-restricted mice. Interestingly, the lipogenic transcription factors and enzymes such as SREBP1c, PPARγ, FASN and ACC were upregulated in the pancreas after CR. In contrast to CR, RYGB decreased the apoptosis of ß-cells and the expression of fatty acid synthase. CONCLUSIONS: Pancreatic fatty acid synthesis is critical to the ß-cell function after CR and RYGB.


Subject(s)
Diabetes Mellitus, Type 2 , Gastric Bypass , Mice , Animals , Diabetes Mellitus, Type 2/metabolism , Caloric Restriction , Pancreas/metabolism , Transcription Factors , Fatty Acids , Lipids
15.
Cells ; 11(20)2022 10 11.
Article in English | MEDLINE | ID: mdl-36291052

ABSTRACT

BACKGROUND: Arrhythmogenic cardiomyopathy (ACM) is a genetic heart muscle disease characterized by progressive fibro-fatty replacement of cardiac myocytes. Up to now, the existing therapeutic modalities for ACM are mostly palliative. About 50% of ACM is caused by mutations in genes encoding desmosomal proteins including Desmoglein-2 (Dsg2). In the current study, the cardiac fibrosis of ACM and its underlying mechanism were investigated by using a cardiac-specific knockout of Dsg2 mouse model. METHODS: Cardiac-specific Dsg2 knockout (CS-Dsg2-/-) mice and wild-type (WT) mice were respectively used as the animal model of ACM and controls. The myocardial collagen volume fraction was determined by histological analysis. The expression levels of fibrotic markers such as α-SMA and Collagen I as well as signal transducers such as STAT3, SMAD3, and PPARα were measured by Western blot and quantitative real-time PCR. RESULTS: Increased cardiac fibrosis was observed in CS-Dsg2-/- mice according to Masson staining. PPARα deficiency and hyperactivation of STAT3 and SMAD3 were observed in the myocardium of CS-Dsg2-/- mice. The biomarkers of fibrosis such as α-SMA and Collagen I were upregulated after gene silencing of Dsg2 in HL-1 cells. Furthermore, STAT3 gene silencing by Stat3 siRNA inhibited the expression of fibrotic markers. The activation of PPARα by fenofibrate or AAV9-Pparα improved the cardiac fibrosis and decreased the phosphorylation of STAT3, SMAD3, and AKT in CS-Dsg2-/- mice. CONCLUSIONS: Activation of PPARα alleviates the cardiac fibrosis in ACM.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia , Desmoglein 2 , Myocardium , PPAR alpha , Animals , Mice , Biomarkers/metabolism , Desmoglein 2/genetics , Desmoglein 2/metabolism , Disease Models, Animal , Fenofibrate/pharmacology , Fibrosis , Myocytes, Cardiac/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/pathology , Myocardium/pathology , Collagen Type I/metabolism
16.
Acta Biochim Biophys Sin (Shanghai) ; 43(7): 501-10, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21642276

ABSTRACT

The mammalian target of rapamycin (mTOR), an evolutionarily conserved serine-threonine protein kinase, belongs to the phosphoinositide 3-kinase (PI3K)-related kinase family, which contains a lipid kinase-like domain within their C-terminal region. Recent studies have revealed that mTOR as a critical intracellular molecule can sense the extracellular energy status and regulate the cell growth and proliferation in a variety of cells and tissues. This review summarizes our current understanding about the effects of mTOR on cell differentiation and tissue development, with an emphasis on the lineage determination of mesenchymal stem cells. mTOR can promote adipogenesis in white adipocytes, brown adipocytes, and muscle satellite cells, while rapamycin inhibits the adipogenic function of mTOR. mTOR signaling may function to affect osteoblast proliferation and differentiation, however, rapamycin has been reported to either inhibit or promote osteogenesis. Although the precise mechanism remains unclear, mTOR is indispensable for myogenesis. Depending on the cell type, rapamycin has been reported to inhibit, promote, or have no effect on myogenesis.


Subject(s)
Cell Differentiation/drug effects , Mesenchymal Stem Cells/physiology , TOR Serine-Threonine Kinases/physiology , Adipogenesis/drug effects , Animals , Cell Lineage , Cell Proliferation/drug effects , Humans , MicroRNAs/physiology , Muscle Development/drug effects , Osteogenesis/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology
17.
Orphanet J Rare Dis ; 16(1): 496, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819141

ABSTRACT

BACKGROUND: The left ventricular noncompaction cardiomyopathy (LVNC) is a rare subtype of cardiomyopathy associated with a high risk of heart failure (HF), thromboembolism, arrhythmia, and sudden cardiac death. METHODS: The proband with overlap phenotypes of LVNC and hypertrophic cardiomyopathy (HCM) complicates atrial fibrillation (AF), ventricular tachycardia (VT), and HF due to the diffuse myocardial lesion, which were diagnosed by electrocardiogram, echocardiogram and cardiac magnetic resonance imaging. Peripheral blood was collected from the proband and his relatives. DNA was extracted from the peripheral blood of proband for high-throughput target capture sequencing. The Sanger sequence verified the variants. The protein was extracted from the skin of the proband and healthy volunteer. The expression difference of desmocollin2 was detected by Western blot. RESULTS: The novel heterozygous truncated mutation (p.K47Rfs*2) of the DSC2 gene encoding an important component of desmosomes was detected by targeted capture sequencing. The western blots showed that the expressing level of functional desmocollin2 protein (~ 94kd) was lower in the proband than that in the healthy volunteer, indicating that DSC2 p.K47Rfs*2 obviously reduced the functional desmocollin2 protein expression in the proband. CONCLUSION: The heterozygous DSC2 p.K47Rfs*2 remarkably and abnormally reduced the functional desmocollin2 expression, which may potentially induce the overlap phenotypes of LVNC and HCM, complicating AF, VT, and HF.


Subject(s)
Cardiomyopathy, Hypertrophic , Heart Failure , Arrhythmias, Cardiac , Cardiomyopathy, Hypertrophic/genetics , Desmocollins/genetics , Heart Failure/genetics , Humans , Mutation/genetics , Phenotype
18.
Cell Physiol Biochem ; 26(4-5): 707-16, 2010.
Article in English | MEDLINE | ID: mdl-21063108

ABSTRACT

BACKGROUND: Ghrelin, the only identified circulating orexigenic signal, is unique in structure in which a specific acyl-modification of its third serine occurs. This acylation is necessary for ghrelin to bind to its receptor and to exert its biologic activity, which is catalyzed by ghrelin O-acyltransferase (GOAT). Although ghrelin is mainly secreted from gastric X/A like endocrine cells, it is also expressed in pancreatic islet cells and regulates insulin secretion. In this study, we examined the expression and regulation of GOAT in pancreas. METHODS: GOAT mRNA and immunoreactivity were examined in pancreatic islets and INS-1 cells by RT-PCR and immunofluorescent staining or Western blotting. RESULTS: Insulin inhibits the expression of GOAT mRNA and GOAT promoter activity in a dose and time-dependent manner. The mammalian target of rapamycin (mTOR) is activated by insulin. Blocking mTOR signaling by either rapamycin or overexpression of its negative regulator tuberous sclerosis complex 1 (TSC1) or TSC2 attenuates the inhibitory effect of insulin on the transcription and translation of GOAT. CONCLUSION: Our study suggests that GOAT is present in pancreatic islet cells and that insulin inhibits the expression of GOAT via the mediation of mTOR signaling.


Subject(s)
Acyltransferases/metabolism , Islets of Langerhans/enzymology , Acyltransferases/genetics , Animals , Anti-Bacterial Agents/pharmacology , Insulin/pharmacology , Male , Mice , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL