1
|
Cadena Sandoval M, Haeusler RA. Bile acid metabolism in type 2 diabetes mellitus. Nat Rev Endocrinol 2025; 21:203-213. [PMID: 39757322 PMCID: PMC12053743 DOI: 10.1038/s41574-024-01067-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/07/2025]
Abstract
Type 2 diabetes mellitus is a complex disorder associated with insulin resistance and hyperinsulinaemia that is insufficient to maintain normal glucose metabolism. Changes in insulin signalling and insulin levels are thought to directly explain many of the metabolic abnormalities that occur in diabetes mellitus, such as impaired glucose disposal. However, molecules that are directly affected by abnormal insulin signalling might subsequently go on to cause secondary metabolic effects that contribute to the pathology of type 2 diabetes mellitus. In the past several years, evidence has linked insulin resistance with the concentration, composition and distribution of bile acids. As bile acids are known to regulate glucose metabolism, lipid metabolism and energy balance, these findings suggest that bile acids are potential mediators of metabolic distress in type 2 diabetes mellitus. In this Review, we highlight advances in our understanding of the complex regulation of bile acids during insulin resistance, as well as how bile acids contribute to metabolic control.
Collapse
Affiliation(s)
- Marti Cadena Sandoval
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA
- Columbia Digestive and Liver Disease Research Center, Columbia University Medical Center, New York, NY, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Columbia Digestive and Liver Disease Research Center, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Kong X, Yan D, Shao L, Li B, Lv S, Tu Y, Zhang Y, Shu X, Ying Y, Ma X. Increased COX6A2 Promotes Pancreatic β-Cell Apoptosis and Is Suppressed in Diabetic GK Rats After Roux-en-Y Gastric Bypass. Diabetes 2025; 74:175-187. [PMID: 39546664 DOI: 10.2337/db24-0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
ARTICLE HIGHLIGHTS Cytochrome c oxidase subunit 6A2 (COX6A2) expression is increased in diabetic islets. Increased COX6A2 promotes β-cell apoptosis via modulation of cyclophilin D-mediated cytochrome c release from mitochondria to the cytoplasm. Carbohydrate-responsive element-binding protein epigenetically regulates COX6A2 expression in β-cells. Roux-en-Y gastric bypass reduces COX6A2 expression by regulating the glucagon-like peptide 1/cAMP-dependent protein kinase/carbohydrate-responsive element-binding protein signaling pathway.
Collapse
Affiliation(s)
- Xiangchen Kong
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Dan Yan
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Lianqi Shao
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Bingfeng Li
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
- Department of Surgery, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Simian Lv
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Yifan Tu
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Yingqi Zhang
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Xingsheng Shu
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Ying Ying
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Xiaosong Ma
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
3
|
Guidi L, Martinez-Tellez B, Ortega Santos CP. Obesity, gut bacteria, and the epigenetic control of metabolic disease. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:333-368. [DOI: 10.1016/b978-0-443-18979-1.00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Shao L, Kong X, Lv S, Shu X, Ma X, Ai X, Yan D, Ying Y. FXR-regulated COX6A2 triggers mitochondrial apoptosis of pancreatic β-cell in type 2 diabetes. Cell Death Dis 2024; 15:920. [PMID: 39702527 DOI: 10.1038/s41419-024-07302-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Pancreatic β-cell apoptosis plays a crucial role in the development of type 2 diabetes. Cytochrome c oxidase subunit 6A2 (COX6A2) and Farnesoid X Receptor (FXR) have been identified in pancreatic β-cells, however, whether they are involved in β-cell apoptosis is unclear. Here, we sought to investigate the role of FXR-regulated COX6A2 in diabetic β-cell apoptosis. We found that COX6A2 expression was increased in islets from diabetic animals, whereas FXR expression was suppressed. Notably, overexpression of COX6A2 facilitated β-cell apoptosis, whereas its deficiency attenuated this process and ameliorates type 2 diabetes, suggesting a pro-apoptotic role of COX6A2 in β-cells. Mechanistically, increased COX6A2 interacted with and enhanced the expression of voltage-dependent anion channel 1 (VDAC1), thereby promoting the mitochondrial translocation of Bax, leading to the release of cytochrome c from the mitochondria to the cytoplasm and ultimately causing β-cell apoptosis. Moreover, FXR negatively regulated COX6A2 expression through the inhibition of histone acetyltransferase p300 occupancy, diminishing histone H3 acetylation at lysine 27 on the Cox6a2 promoter. Furthermore, the deficiency of FXR intensified β-cell apoptosis under diabetic situations. Thus, it is probable that in diabetogenic environments, reduced FXR expression contributes to enhanced COX6A2 expression, culminating in β-cell apoptosis. These findings emphasize the essential involvement of the FXR/p300 pathway-controlled COX6A2 in β-cell apoptosis, revealing a previously undiscovered mechanism underlying diabetic β-cell apoptosis.
Collapse
Affiliation(s)
- Lianqi Shao
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China
- Guangdong Key Laboratory for Biomedical Measurements and U1trasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical U1trasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, PR China
| | - Xiangchen Kong
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China.
| | - Simian Lv
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China
- School of Basic Medicine, Jiamusi University, Jiamusi, PR China
| | - Xingsheng Shu
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China
| | - Xiaosong Ma
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China
| | - Xiaojiao Ai
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China
| | - Dan Yan
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China.
| | - Ying Ying
- Diabetes Institute, the Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis ZDSYS, Shenzhen University Medical School, Shenzhen, PR China.
| |
Collapse
|
5
|
Hu A, Lan H, Yao Z, Kong X. Carbohydrate response element-binding protein (ChREBP) mediates decreased SNAP25 expression in islets from diabetic Goto-Kakizaki (GK) rats. FEBS Open Bio 2024; 14:1864-1872. [PMID: 39300600 PMCID: PMC11532970 DOI: 10.1002/2211-5463.13900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/15/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
SNAP25 plays an essential role in the glucose-stimulated insulin secretion (GSIS) of pancreatic β-cells. Carbohydrate response element-binding protein (ChREBP) is an important transcription factor in β-cells and, in this study, we aimed to explore whether ChREBP regulates SNAP25 expression in β-cells. We show that diabetic Goto-Kakizaki (GK) rats exhibited impaired insulin secretion and hyperglycemia, along with decreased SNAP25 expression and ChREBP phosphorylation in islets. SNAP25 knockdown decreased GSIS in β-cells, while SNAP25 overexpression increased GSIS in β-cells. Activation or overexpression of ChREBP led to reduced SNAP25 expression and subsequent suppression of GSIS. Conversely, ChREBP knockdown mitigated the reduction in SNAP25 expression caused by high glucose. Mechanistically, the activation of ChREBP by high glucose increased its occupancy and decreased the level of H3K4me3 at the Snap25 promoter. Our findings reveal the novel regulatory mechanisms of SNAP25 expression in β-cells and suggest that SNAP25 may be involved in the regulation of β-cell secretory function controlled by ChREBP.
Collapse
Affiliation(s)
- Anyi Hu
- Shenzhen University Diabetes Institute, School of MedicineShenzhen UniversityChina
| | - Hongyan Lan
- Shenzhen University Diabetes Institute, School of MedicineShenzhen UniversityChina
| | - Zilai Yao
- Shenzhen University Diabetes Institute, School of MedicineShenzhen UniversityChina
| | - Xiangchen Kong
- Shenzhen University Diabetes Institute, School of MedicineShenzhen UniversityChina
| |
Collapse
|
6
|
Yang D. TRPA1-Related Diseases and Applications of Nanotherapy. Int J Mol Sci 2024; 25:9234. [PMID: 39273183 PMCID: PMC11395144 DOI: 10.3390/ijms25179234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Transient receptor potential (TRP) channels, first identified in Drosophila in 1969, are multifunctional ion channels expressed in various cell types. Structurally, TRP channels consist of six membrane segments and are classified into seven subfamilies. Transient receptor potential ankyrin 1 (TRPA1), the first member of the TRPA family, is a calcium ion affinity non-selective cation channel involved in sensory transduction and responds to odors, tastes, and chemicals. It also regulates temperature and responses to stimuli. Recent studies have linked TRPA1 to several disorders, including chronic pain, inflammatory diseases, allergies, and respiratory problems, owing to its activation by environmental toxins. Mutations in TRPA1 can affect the sensory nerves and microvasculature, potentially causing nerve pain and vascular problems. Understanding the function of TRPA1 is important for the development of treatments for these diseases. Recent developments in nanomedicines that target various ion channels, including TRPA1, have had a significant impact on disease treatment, providing innovative alternatives to traditional disease treatments by overcoming various adverse effects.
Collapse
Affiliation(s)
- Dongki Yang
- Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
7
|
Liu Z, Chen L, Chen M, Linghu L, Liao Z, Chen M, Wang G. Sarmentol H derived from Sedum sarmentosum Bunge directly targets FXR to mitigate cholestasis by recruiting SRC-1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155759. [PMID: 38788394 DOI: 10.1016/j.phymed.2024.155759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/17/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Farnesoid X receptor (FXR) is a vital receptor for bile acids and plays an important role in the treatment of cholestatic liver disease. In addition to traditional bile acid-based steroidal agonists, synthetic alkaloids are the most commonly reported non-steroidal FXR agonists. Sarmentol H is a nor-sesquiterpenoid obtained from Sedum sarmentosum Bunge, and in vitro screening experiments have shown that it might be related to the regulation of the FXR pathway in a previous study. PURPOSE To investigate the therapeutic effects of sarmentol H on cholestasis and to determine whether sarmentol H directly targets FXR to mitigate cholestasis. Furthermore, this study aimed to explore the key amino acid residues involved in the binding of sarmentol H to FXR through site-directed mutagenesis. METHODS An intrahepatic cholestasis mouse model was established to investigate the therapeutic effects of sarmentol H on cholestasis. In vitro experiments, including Co-Ip and FXR-EcRE-Luc assays, were performed to assess whether sarmentol H activates FXR by recruiting the receptor coactivator SRC1. CETSA, SIP, DARTS, and ITC were used to determine the binding of sarmentol H to FXR protein. The key amino acid residues for sarmentol H binding to FXR were analyzed by molecular docking and site-directed mutagenesis. Finally, we conducted in vivo experiments on wild-type and Fxr-/- mice to further validate the anticholestatic target of sarmentol H. RESULTS Sarmentol H had significant ameliorative effects on the pathological conditions of cholestatic mice induced with ANIT. In vitro experiments suggested that it is capable of activating FXR and regulating downstream signaling pathways by recruiting SRC1. The target validation experiments showed that sarmentol H had the ability to bind to FXR as a ligand (KD = 2.55 μmol/L) and enhance the stability of its spatial structure. Moreover, site-directed mutagenesis revealed that THR292 and TYR365 were key binding sites for sarmentol H and FXR. Furthermore, knockout of the Fxr gene resulted in a significantly higher degree of ANIT-induced cholestatic liver injury than that in wild-type cholestatic mice, and the amelioration of cholestasis or regulatory effects on FXR downstream genes by sarmentol H also disappeared in Fxr-/- cholestatic mice. CONCLUSION Sarmentol H is an FXR agonist. This is the first study to show that it exerts a significant therapeutic effect on cholestatic mice, and can directly bind to FXR and activate it by recruiting the coactivator SRC1.
Collapse
Affiliation(s)
- Zhenxiu Liu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Lin Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Mingyun Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Lang Linghu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Zhihua Liao
- School of Life Sciences, Southwest University, Chongqing, China
| | - Min Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| | - Guowei Wang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| |
Collapse
|
8
|
Kong X, Yang C, Li B, Yan D, Yang Y, Cao C, Xing B, Ma X. FXR/Menin-mediated epigenetic regulation of E2F3 expression controls β-cell proliferation and is increased in islets from diabetic GK rats after RYGB. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167136. [PMID: 38531483 DOI: 10.1016/j.bbadis.2024.167136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
Farnesoid X receptor (FXR) improves the function of islets, especially in the setting of Roux-en-Y gastric bypass (RYGB). Here we investigated how FXR activation regulates β-cell proliferation and explored the potential link between FXR signaling and the menin pathway in controlling E2F3 expression, a key transcription factor for controlling adult β-cell proliferation. Stimulation with the FXR agonist GW4064 or chenodeoxycholic acid (CDCA) increased E2F3 expression and β-cell proliferation. Consistently, E2F3 knockdown abolished GW4064-induced proliferation. Treatment with GW4064 increased E2F3 expression in β-cells via enhancing Steroid receptor coactivator-1 (SRC1) recruitment, increasing the pro-transcriptional acetylation of histone H3 at the E2f3 promoter. GW4064 treatment also decreased the association between FXR and menin, leading to the induction of FXR-mediated SRC1 recruitment. Mimicking the impact of FXR agonists, RYGB also increased E2F3 expression and β-cell proliferation in GK rats and SD rats. These findings unravel the crucial role of the FXR/menin signaling in epigenetically controlling E2F3 expression and β-cell proliferation, a mechanism possibly underlying RYGB-induced β-cell proliferation.
Collapse
Affiliation(s)
- Xiangchen Kong
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Chenxi Yang
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Bingfeng Li
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Dan Yan
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Yanhui Yang
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Cuihua Cao
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Bowen Xing
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Xiaosong Ma
- Shenzhen University Diabetes Institute, School of Medicine, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
9
|
Kong X, Feng L, Yan D, Li B, Yang Y, Ma X. FXR-mediated epigenetic regulation of GLP-1R expression contributes to enhanced incretin effect in diabetes after RYGB. J Cell Mol Med 2024; 28:e16339. [PMID: 33611845 PMCID: PMC10941525 DOI: 10.1111/jcmm.16339] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 01/07/2023] Open
Abstract
In this study, we investigated how Roux-en-Y gastric bypass (RYGB) enhances glucagon-like peptide 1 (GLP-1) response in GK rats and explored the potential link between RYGB-stimulated BAs/FXR signalling and GLP-1R-linked signalling in β-cells, a key pathway that regulates glucose-stimulated insulin secretion (GSIS). Here we show that RYGB restores GLP-1R expression in GK rat islets. This involves increased total BAs as well as chenodeoxycholic acid (CDCA), leading to FXR activation, increasing FXR binding to the promoter of Glp-1r and enhancing occupancy of histone acetyltransferase steroid receptor coactivator-1 (SRC1), thus increasing histone H3 acetylation at the promoter. These coordinated events bring about increased GLP-1R expression, resulting in greater GLP-1 response in β-cells. Moreover, ablation of FXR suppressed the stimulatory effects of GLP-1. Thus, this study unravels the crucial role of the BAs/FXR/SRC1 axis-controlled GLP-1R expression in β-cells, which results in enhanced incretin effect and normalized blood glucose of GK rats after RYGB.
Collapse
Affiliation(s)
- Xiangchen Kong
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Linxian Feng
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Dan Yan
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Bingfeng Li
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Yanhui Yang
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Xiaosong Ma
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| |
Collapse
|
10
|
Yan D, Lv M, Kong X, Feng L, Ying Y, Liu W, Wang X, Ma X. FXR controls insulin content by regulating Foxa2-mediated insulin transcription. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119655. [PMID: 38135007 DOI: 10.1016/j.bbamcr.2023.119655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Farnesoid X receptor (FXR) is a nuclear ligand-activated receptor of bile acids that plays a role in the modulation of insulin content. However, the underlying molecular mechanisms remain unclear. Forkhead box a2 (Foxa2) is an important nuclear transcription factor in pancreatic β-cells and is involved in β-cell function. We aimed to explore the signaling mechanism downstream of FXR to regulate insulin content and underscore its association with Foxa2 and insulin gene (Ins) transcription. All experiments were conducted on FXR transgenic mice, INS-1 823/13 cells, and diabetic Goto-Kakizaki (GK) rats undergoing sham or Roux-en-Y gastric bypass (RYGB) surgery. Islets from FXR knockout mice and INS-1823/13 cells with FXR knockdown exhibited substantially lower insulin levels than that of controls. This was accompanied by decreased Foxa2 expression and Ins transcription. Conversely, FXR overexpression increased insulin content, concomitant with enhanced Foxa2 expression and Ins transcription in INS-1 823/13 cells. Moreover, FXR knockdown reduced FXR recruitment and H3K27 trimethylation in the Foxa2 promoter. Importantly, Foxa2 overexpression abrogated the adverse effects of FXR knockdown on Ins transcription and insulin content in INS-1 823/13 cells. Notably, RYGB surgery led to improved insulin content in diabetic GK rats, which was accompanied by upregulated FXR and Foxa2 expression and Ins transcription. Collectively, these data suggest that Foxa2 serves as the target gene of FXR in β-cells and mediates FXR-enhanced Ins transcription. Additionally, the upregulated FXR/Foxa2 signaling cascade could contribute to the enhanced insulin content in diabetic GK rats after RYGB.
Collapse
Affiliation(s)
- Dan Yan
- Shenzhen University Diabetes Institute, Medical School, Shenzhen University, Shenzhen 518060, PR China.
| | - Moyang Lv
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, PR China
| | - Xiangchen Kong
- Shenzhen University Diabetes Institute, Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Linxian Feng
- Shenzhen University Diabetes Institute, Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Ying Ying
- Shenzhen University Diabetes Institute, Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Wenjuan Liu
- Shenzhen University Diabetes Institute, Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Xin Wang
- Shenzhen University Diabetes Institute, Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Xiaosong Ma
- Shenzhen University Diabetes Institute, Medical School, Shenzhen University, Shenzhen 518060, PR China
| |
Collapse
|
11
|
Kong X, Lin C, Yang C, Wang X, Li B, Yan D, Yang Y, Hu A, Chen Y, Xu X, Ma X. GLP-1 signaling-regulated SNAP25 is involved in improved insulin secretion in diabetic GK rats after Roux-en-Y gastric bypass surgery. Mol Biol Rep 2024; 51:123. [PMID: 38227062 DOI: 10.1007/s11033-023-09165-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Roux-en-Y gastric bypass surgery (RYGB) improves glucose-stimulated insulin secretion (GSIS) in type 2 diabetes (T2D) patients. SNAP25 plays an essential role in GSIS. Clinical studies indicate that enhanced GLP-1 signaling is an important contributor to the improved β-cell function in T2D. We aimed to explore whether GLP-1-regulated SNAP25 is involved in the enhanced secretory function of β-cells in diabetic Goto-Kakizaki (GK) rats after RYGB. METHODS AND RESULTS RYGB or sham surgery was conducted in GK rats. mRNA and protein expression of SNAP25 was assessed by qPCR and Western blot, respectively. Occupancy of CREB and acetyltransferase CBP and acetylation of histone H3 (ACH3) at the Snap25 promoter were determined using ChIP assay. RYGB led to increased SNAP25 expression and CREB phosphorylation in islets from GK rats. Increased SNAP25 improved GSIS in β-cells cultured in high glucose conditions. Consistent with increased plasma GLP-1 after RYGB, GLP-1R agonist exendin4 increased SNAP25 expression and CREB phosphorylation in β-cells. Mechanistically, exendin4 promoted the recruitment of CREB and CBP, thereby increasing ACH3 at the Snap25 promoter. Consistently, inhibition of CBP attenuated the effect of exendin4 on SNAP25 expression. Furthermore, the knockdown of SNAP25 diminished the increase of GSIS potentiated by chronic GLP-1 culture in INS-1 832/13 cells. CONCLUSIONS Our findings unravel the novel mechanisms of RYGB-enhanced SNAP25 expression in β-cells, and SNAP25 may contribute to the improved β-cell secretory function induced by RYGB.
Collapse
Affiliation(s)
- Xiangchen Kong
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China.
| | - Chao Lin
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Chenxi Yang
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Xin Wang
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Bingfeng Li
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Dan Yan
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Yanhui Yang
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Anyi Hu
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Yanyin Chen
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| | - Xiaohui Xu
- Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Xiaosong Ma
- School of Medicine, Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
12
|
Ferreira FP, Pereira SS, Costa MM, Guimarães M, Albrechtsen NJW, Holst JJ, Nora M, Monteiro MP. Individuals with type 2 diabetes have higher density of small intestinal neurotensin-expressing cells. Mol Cell Biochem 2023; 478:2779-2787. [PMID: 36920577 PMCID: PMC10627918 DOI: 10.1007/s11010-023-04698-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 03/01/2023] [Indexed: 03/16/2023]
Abstract
Neurotensin (NT) is a gastro-intestinal hormone involved in several pathways that regulate energy and glucose homeostasis. NT was hypothesized to act in synergy with incretin hormones to potentiate its anti-diabetic effects. Additionally, circulating NT levels were shown to rise after bariatric surgery-induced weight loss. Knowledge of NT-secreting cells distribution along the small intestine and its variation according to diabetes status could provide insights on NT role in mediating type 2 diabetes (T2D) improvement after bariatric surgery. So, our aims were to characterize NT-expressing cell distribution along the human small intestine and to compare the relative density of NT-expressing cells in the small intestine of individuals with and without T2D undergoing bariatric surgery for obesity treatment. Autopsy-derived small intestine fragments (n = 30) were obtained at every 20 cm along the entire intestinal length. Additionally, jejunum biopsies (n = 29) were obtained during elective gastric bypass interventions from patients with (n = 10) or without T2D (n = 18). NT-expressing cells were identified by immunohistochemistry and quantified via computerized morphometric analysis. NT-expressing cell density increased along the human small intestine. NT-expressing cell density was significantly higher from 200 cm distal to the duodenojejunal flexure onward, as well as in subjects with T2D when compared to those without T2D. NT-expressing cell density increases along the human small gut, and a higher density is found in individuals with T2D. This finding suggests a potential role for NT in the mechanisms of disease and T2D improvement observed after bariatric surgery.
Collapse
Affiliation(s)
- Filipa P Ferreira
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Sofia S Pereira
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal.
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Madalena M Costa
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Marta Guimarães
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre Douro E Vouga, Santa Maria da Feira, Portugal
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2100, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Mário Nora
- Department of General Surgery, Centro Hospitalar de Entre Douro E Vouga, Santa Maria da Feira, Portugal
| | - Mariana P Monteiro
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| |
Collapse
|
13
|
Wu W, Zheng J, Wang R, Wang Y. Ion channels regulate energy homeostasis and the progression of metabolic disorders: Novel mechanisms and pharmacology of their modulators. Biochem Pharmacol 2023; 218:115863. [PMID: 37863328 DOI: 10.1016/j.bcp.2023.115863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
The progression of metabolic diseases, featured by dysregulated metabolic signaling pathways, is orchestrated by numerous signaling networks. Among the regulators, ion channels transport ions across the membranes and trigger downstream signaling transduction. They critically regulate energy homeostasis and pathogenesis of metabolic diseases and are potential therapeutic targets for treating metabolic disorders. Ion channel blockers have been used to treat diabetes for decades by stimulating insulin secretion, yet with hypoglycemia and other adverse effects. It calls for deeper understanding of the largely elusive regulatory mechanisms, which facilitates the identification of new therapeutic targets and safe drugs against ion channels. In the article, we critically assess the two principal regulatory mechanisms, protein-channel interaction and post-translational modification on the activities of ion channels to modulate energy homeostasis and metabolic disorders through multiple novel mechanisms. Moreover, we discuss the multidisciplinary methods that provide the tools for elucidation of the regulatory mechanisms mediating metabolic disorders by ion channels. In terms of translational perspective, the mechanistic analysis of recently validated ion channels that regulate insulin resistance, body weight control, and adverse effects of current ion channel antagonists are discussed in details. Their small molecule modulators serve as promising new drug candidates to combat metabolic disorders.
Collapse
Affiliation(s)
- Wenyi Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Jianan Zheng
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, China
| | - Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, China.
| |
Collapse
|
14
|
Wen S, Gong M, Wang T, Zhou M, Dong M, Li Y, Xu C, Yuan Y, Zhou L. The Rapid Changes in Bodyweight and Glycemic Control Are Determined by Pre-status After Bariatric Surgery in Both Genders in Young Chinese Individuals. Cureus 2023; 15:e46603. [PMID: 37937018 PMCID: PMC10626214 DOI: 10.7759/cureus.46603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
PURPOSES The primary aim of this clinical study is to identify the factors associated with rapid glycemic, bodyweight, and lipid profile remission in young obese patients following bariatric surgery. MATERIALS AND METHODS In a total of 131 Chinese in-patients at Shanghai Pudong Hospital, China, we retrospectively analyzed in-patient data of metabolic parameters, including BMI, waist circumference, blood pressure (BP), and blood laboratory tests, such as plasma lipids and lipoprotein, hemoglobulin A1c (HbA1c), and oral glucose tolerance tests (OGTT) before bariatric surgery. We followed up these indices at the first month, third months, half-year, and one year later. RESULTS The results showed that bodyweight, BP, fasting plasma glucose (FPG), HbA1c, and triglyceride (TG) levels decreased significantly in one to three months following surgery in both male and female patients (p<0.05). We demonstrated that age (male: β=-0.181; female: β=-0.292) and the pre-operation HbA1c levels (male: β=0.935; female: β=0.919) were independent predictors of HbA1c reduction in both young obese male and female patients in three months after surgery. For body weight loss, age (β=-0.229) and pre-operation bodyweight (β=0.735) are the predictors in females, but only pre-operation body weight (β=0.798) is the independent predictor in obese young male patients. CONCLUSION This study discovered that changes in bodyweight were determined by age, pre-operation status of bodyweight, and HbA1C in obese young Chinese.
Collapse
Affiliation(s)
- Song Wen
- Endocrinology, Shanghai Pudong Hospital, Shanghai, CHN
| | - Min Gong
- Endocrinology, Shanghai Pudong Hospital, Shanghai, CHN
| | | | - Mingyue Zhou
- Gynecology, University of California San Francisco, San Francisco, USA
| | - Meiyuan Dong
- Endocrinology, Shanghai Pudong Hospital, Shanghai, CHN
| | - Yanyan Li
- Endocrinology, Shanghai Pudong Hospital, Shanghai, CHN
| | - Chenglin Xu
- Endocrinology, Shanghai Pudong Hospital, Shanghai, CHN
| | - Yue Yuan
- Endocrinology, Shanghai Pudong Hospital, Shanghai, CHN
| | - Ligang Zhou
- Endocrinology, Shanghai Pudong Hospital, Shanghai, CHN
| |
Collapse
|
15
|
Fila M, Pawlowska E, Szczepanska J, Blasiak J. Epigenetic Connections of the TRPA1 Ion Channel in Pain Transmission and Neurogenic Inflammation - a Therapeutic Perspective in Migraine? Mol Neurobiol 2023; 60:5578-5591. [PMID: 37326902 PMCID: PMC10471718 DOI: 10.1007/s12035-023-03428-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
Persistent reprogramming of epigenetic pattern leads to changes in gene expression observed in many neurological disorders. Transient receptor potential cation channel subfamily A member 1 (TRPA1), a member of the TRP channels superfamily, is activated by many migraine triggers and expressed in trigeminal neurons and brain regions that are important in migraine pathogenesis. TRP channels change noxious stimuli into pain signals with the involvement of epigenetic regulation. The expression of the TRPA1 encoding gene, TRPA1, is modulated in pain-related syndromes by epigenetic alterations, including DNA methylation, histone modifications, and effects of non-coding RNAs: micro RNAs (miRNAs), long non-coding RNAs, and circular RNAs. TRPA1 may change epigenetic profile of many pain-related genes as it may modify enzymes responsible for epigenetic modifications and expression of non-coding RNAs. TRPA1 may induce the release of calcitonin gene related peptide (CGRP), from trigeminal neurons and dural tissue. Therefore, epigenetic regulation of TRPA1 may play a role in efficacy and safety of anti-migraine therapies targeting TRP channels and CGRP. TRPA1 is also involved in neurogenic inflammation, important in migraine pathogenesis. The fundamental role of TRPA1 in inflammatory pain transmission may be epigenetically regulated. In conclusion, epigenetic connections of TRPA1 may play a role in efficacy and safety of anti-migraine therapy targeting TRP channels or CGRP and they should be further explored for efficient and safe antimigraine treatment. This narrative/perspective review presents information on the structure and functions of TRPA1 as well as role of its epigenetic connections in pain transmission and potential in migraine therapy.
Collapse
Affiliation(s)
- Michal Fila
- Department of Developmental Neurology and Epileptology, Polish Mother's Memorial Hospital Research Institute, 93-338, Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-217, Lodz, Poland
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-217, Lodz, Poland
| | - Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, 90-236, Lodz, Poland.
| |
Collapse
|
16
|
Abstract
Many people with obesity and type 2 diabetes achieve remission of their diabetes after Roux-en-Y Gastric Bypass Surgery but the mechanisms remain disputed. We provide our perspective on competing data sets that either point towards this effect being entirely due to the loss of weight, or due to weight loss-independent effects.
Collapse
Affiliation(s)
- Samuel Klein
- Center for Human Nutrition, Washington University, St. Louis, Missouri, USA
- Sansum Diabetes Research Institute, Santa Barbara, CA, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
17
|
Wang Y. Multidisciplinary Advances Address the Challenges in Developing Drugs against Transient Receptor Potential Channels to Treat Metabolic Disorders. ChemMedChem 2023; 18:e202200562. [PMID: 36530131 DOI: 10.1002/cmdc.202200562] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/01/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Transient receptor potential (TRP) channels are cation channels that regulate key physiological and pathological processes in response to a broad range of stimuli. Moreover, they systemically regulate the release of hormones, metabolic homeostasis, and complications of diabetes, which positions them as promising therapeutic targets to combat metabolic disorders. Nevertheless, there are significant challenges in the design of TRP ligands with high potency and durability. Herein we summarize the four challenges as hydrophobicity, selectivity, mono-target therapy, and interspecies discrepancy. We present 1134 TRP ligands with diversified modes of TRP-ligand interaction and provide a detailed discussion of the latest strategies, especially cryogenic electron microscopy (cryo-EM) and computational methods. We propose solutions to address the challenges with a critical analysis of advances in membrane partitioning, polypharmacology, biased agonism, and biochemical screening of transcriptional modulators. They are fueled by the breakthrough from cryo-EM, chemoinformatics and bioinformatics. The discussion is aimed to shed new light on designing next-generation drugs to treat obesity, diabetes and its complications, with optimal hydrophobicity, higher mode selectivity, multi-targeting and consistent activities between human and rodents.
Collapse
Affiliation(s)
- Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, P. R. China.,Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, 200438, P. R. China
| |
Collapse
|
18
|
Guo Y, Xie G, Zhang X. Role of FXR in Renal Physiology and Kidney Diseases. Int J Mol Sci 2023; 24:2408. [PMID: 36768731 PMCID: PMC9916923 DOI: 10.3390/ijms24032408] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Farnesoid X receptor, also known as the bile acid receptor, belongs to the nuclear receptor (NR) superfamily of ligand-regulated transcription factors, which performs its functions by regulating the transcription of target genes. FXR is highly expressed in the liver, small intestine, kidney and adrenal gland, maintaining homeostasis of bile acid, glucose and lipids by regulating a diverse array of target genes. It also participates in several pathophysiological processes, such as inflammation, immune responses and fibrosis. The kidney is a key organ that manages water and solute homeostasis for the whole body, and kidney injury or dysfunction is associated with high morbidity and mortality. In the kidney, FXR plays an important role in renal water reabsorption and is thought to perform protective functions in acute kidney disease and chronic kidney disease, especially diabetic kidney disease. In this review, we summarize the recent advances in the understanding of the physiological and pathophysiological function of FXR in the kidney.
Collapse
Affiliation(s)
| | | | - Xiaoyan Zhang
- Health Science Center, East China Normal University, Shanghai 200241, China
| |
Collapse
|
19
|
Zhou W, Anakk S. Enterohepatic and non-canonical roles of farnesoid X receptor in controlling lipid and glucose metabolism. Mol Cell Endocrinol 2022; 549:111616. [PMID: 35304191 PMCID: PMC9245558 DOI: 10.1016/j.mce.2022.111616] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Farnesoid X receptor (FXR) is a nuclear receptor that transcriptionally regulates bile acid homeostasis along with nutrient metabolism. In addition to the gastrointestinal (GI) tract, FXR expression has been widely noted in kidney, adrenal gland, pancreas, adipose, skeletal muscle, heart, and brain. Except for the liver and gut, the relevance of FXR signaling in metabolism in other tissues remains poorly understood. This review examines the classical and non-canonical tissue-specific roles of FXR in regulating, lipids, and glucose homeostasis under normal and diseased states. FXR activation has been reported to be protective against cholestasis, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), type 2 diabetes, cardiovascular and kidney diseases. Several ongoing clinical trials are investigating FXR ligands as a therapeutic target for primary biliary cholangitis (PBC) and NASH, which substantiate the significance of FXR signaling in modulating metabolic processes. This review highlights that FXR ligands, albeit an attractive therapeutic target for treating metabolic diseases, tissue-specific modulation of FXR may be the key to overcoming some of the adverse clinical effects.
Collapse
Affiliation(s)
- Weinan Zhou
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
20
|
Cotero V, Graf J, Miwa H, Hirschstein Z, Qanud K, Huerta TS, Tai N, Ding Y, Jimenez-Cowell K, Tomaio JN, Song W, Devarajan A, Tsaava T, Madhavan R, Wallace K, Loghin E, Morton C, Fan Y, Kao TJ, Akhtar K, Damaraju M, Barenboim L, Maietta T, Ashe J, Tracey KJ, Coleman TR, Di Carlo D, Shin D, Zanos S, Chavan SS, Herzog RI, Puleo C. Stimulation of the hepatoportal nerve plexus with focused ultrasound restores glucose homoeostasis in diabetic mice, rats and swine. Nat Biomed Eng 2022; 6:683-705. [PMID: 35361935 PMCID: PMC10127248 DOI: 10.1038/s41551-022-00870-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
Abstract
Peripheral neurons that sense glucose relay signals of glucose availability to integrative clusters of neurons in the brain. However, the roles of such signalling pathways in the maintenance of glucose homoeostasis and their contribution to disease are unknown. Here we show that the selective activation of the nerve plexus of the hepatic portal system via peripheral focused ultrasound stimulation (pFUS) improves glucose homoeostasis in mice and rats with insulin-resistant diabetes and in swine subject to hyperinsulinemic-euglycaemic clamps. pFUS modulated the activity of sensory projections to the hypothalamus, altered the concentrations of metabolism-regulating neurotransmitters, and enhanced glucose tolerance and utilization in the three species, whereas physical transection or chemical blocking of the liver-brain nerve pathway abolished the effect of pFUS on glucose tolerance. Longitudinal multi-omic profiling of metabolic tissues from the treated animals confirmed pFUS-induced modifications of key metabolic functions in liver, pancreas, muscle, adipose, kidney and intestinal tissues. Non-invasive ultrasound activation of afferent autonomic nerves may represent a non-pharmacologic therapy for the restoration of glucose homoeostasis in type-2 diabetes and other metabolic diseases.
Collapse
Affiliation(s)
- Victoria Cotero
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - John Graf
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Hiromi Miwa
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Khaled Qanud
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Tomás S Huerta
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Yuyan Ding
- Yale School of Medicine, New Haven, CT, USA
| | - Kevin Jimenez-Cowell
- Yale School of Medicine, New Haven, CT, USA
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Weiguo Song
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Alex Devarajan
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Tea Tsaava
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Radhika Madhavan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Kirk Wallace
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Evelina Loghin
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Christine Morton
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Ying Fan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Tzu-Jen Kao
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | | | | | | | | | - Jeffrey Ashe
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Kevin J Tracey
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Dino Di Carlo
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Stavros Zanos
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | | | - Chris Puleo
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA.
| |
Collapse
|
21
|
Yoshihara E. Adapting Physiology in Functional Human Islet Organogenesis. Front Cell Dev Biol 2022; 10:854604. [PMID: 35557947 PMCID: PMC9086403 DOI: 10.3389/fcell.2022.854604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023] Open
Abstract
Generation of three-dimensional (3D)-structured functional human islets is expected to be an alternative cell source for cadaveric human islet transplantation for the treatment of insulin-dependent diabetes. Human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer infinite resources for newly synthesized human islets. Recent advancements in hPSCs technology have enabled direct differentiation to human islet-like clusters, which can sense glucose and secrete insulin, and those islet clusters can ameliorate diabetes when transplanted into rodents or non-human primates (NHPs). However, the generated hPSC-derived human islet-like clusters are functionally immature compared with primary human islets. There remains a challenge to establish a technology to create fully functional human islets in vitro, which are functionally and transcriptionally indistinguishable from cadaveric human islets. Understanding the complex differentiation and maturation pathway is necessary to generate fully functional human islets for a tremendous supply of high-quality human islets with less batch-to-batch difference for millions of patients. In this review, I summarized the current progress in the generation of 3D-structured human islets from pluripotent stem cells and discussed the importance of adapting physiology for in vitro functional human islet organogenesis and possible improvements with environmental cues.
Collapse
Affiliation(s)
- Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
22
|
Araújo MC, Soczek SHS, Pontes JP, Marques LAC, Santos GS, Simão G, Bueno LR, Maria-Ferreira D, Muscará MN, Fernandes ES. An Overview of the TRP-Oxidative Stress Axis in Metabolic Syndrome: Insights for Novel Therapeutic Approaches. Cells 2022; 11:cells11081292. [PMID: 35455971 PMCID: PMC9030853 DOI: 10.3390/cells11081292] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/19/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome (MS) is a complex pathology characterized by visceral adiposity, insulin resistance, arterial hypertension, and dyslipidaemia. It has become a global epidemic associated with increased consumption of high-calorie, low-fibre food and sedentary habits. Some of its underlying mechanisms have been identified, with hypoadiponectinemia, inflammation and oxidative stress as important factors for MS establishment and progression. Alterations in adipokine levels may favour glucotoxicity and lipotoxicity which, in turn, contribute to inflammation and cellular stress responses within the adipose, pancreatic and liver tissues, in addition to hepatic steatosis. The multiple mechanisms of MS make its clinical management difficult, involving both non-pharmacological and pharmacological interventions. Transient receptor potential (TRP) channels are non-selective calcium channels involved in a plethora of physiological events, including energy balance, inflammation and oxidative stress. Evidence from animal models of disease has contributed to identify their specific contributions to MS and may help to tailor clinical trials for the disease. In this context, the oxidative stress sensors TRPV1, TRPA1 and TRPC5, play major roles in regulating inflammatory responses, thermogenesis and energy expenditure. Here, the interplay between these TRP channels and oxidative stress in MS is discussed in the light of novel therapies to treat this syndrome.
Collapse
Affiliation(s)
- Mizael C. Araújo
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Suzany H. S. Soczek
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Jaqueline P. Pontes
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 565085-080, MA, Brazil;
| | - Leonardo A. C. Marques
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Gabriela S. Santos
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Laryssa R. Bueno
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Marcelo N. Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Elizabeth S. Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
- Correspondence:
| |
Collapse
|
23
|
Tu J, Wang Y, Jin L, Huang W. Bile acids, gut microbiota and metabolic surgery. Front Endocrinol (Lausanne) 2022; 13:929530. [PMID: 36072923 PMCID: PMC9441571 DOI: 10.3389/fendo.2022.929530] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic surgery, or bariatric surgery, is currently the most effective approach for treating obesity and its complications. Vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass (RYGB) are the top two types of commonly performed metabolic surgery now. The precise mechanisms of how the surgeries work are still unclear, therefore much research has been conducted in this area. Gut hormones such as GLP-1 and PYY have been studied extensively in the context of metabolic surgery because they both participate in satiety and glucose homeostasis. Bile acids, whose functions cover intestinal lipid absorption and various aspects of metabolic regulation via the action of FXR, TGR5, and other bile acid receptors, have also been actively investigated as potential mediators of metabolic surgery. Additionally, gut microbiota and their metabolites have also been studied because they can affect metabolic health. The current review summarizes and compares the recent scientific progress made on identifying the mechanisms of RYGB and VSG. One of the long-term goals of metabolic/bariatric surgery research is to develop new pharmacotherapeutic options for the treatment of obesity and diabetes. Because obesity is a growing health concern worldwide, there is a dire need in developing novel non-invasive treatment options.
Collapse
Affiliation(s)
- Jui Tu
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Irell & Manella Graduate School of Biomedical Science, City of Hope National Medical Center, Duarte, CA, United States
| | - Yangmeng Wang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Lihua Jin
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Irell & Manella Graduate School of Biomedical Science, City of Hope National Medical Center, Duarte, CA, United States
- *Correspondence: Wendong Huang,
| |
Collapse
|
24
|
Mahajan N, Khare P, Kondepudi KK, Bishnoi M. TRPA1: Pharmacology, natural activators and role in obesity prevention. Eur J Pharmacol 2021; 912:174553. [PMID: 34627805 DOI: 10.1016/j.ejphar.2021.174553] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/26/2022]
Abstract
Transient receptor potential ankyrin 1 (TRPA1) channel is a calcium permeable, non-selective cation channel, expressed in the sensory neurons and non-neuronal cells of different tissues. Initially studied for its role in pain and inflammation, TRPA1 has now functionally involved in multiple other physiological functions. TRPA1 channel has been extensively studied for modulation by pungent compounds present in the spices and herbs. In the last decade, the role of TRPA1 agonism in body weight reduction, secretion of hunger and satiety hormones, insulin secretion and thermogenesis, has unveiled the potential of the TRPA1 channel to be used as a preventive target to tackle obesity and associated comorbidities including insulin resistance in type 2 diabetes. In this review, we summarized the recent findings of TRPA1 based dietary/non-dietary modulation for its role in obesity prevention and therapeutics.
Collapse
Affiliation(s)
- Neha Mahajan
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India; Regional Centre for Biotechnology, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Pragyanshu Khare
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India
| | - Kanthi Kiran Kondepudi
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India
| | - Mahendra Bishnoi
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India.
| |
Collapse
|
25
|
Xue H, Huang L, Tu J, Ding L, Huang W. Bile acids and metabolic surgery. LIVER RESEARCH 2021; 5:164-170. [PMID: 39957846 PMCID: PMC11791848 DOI: 10.1016/j.livres.2021.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/04/2021] [Accepted: 05/05/2021] [Indexed: 02/16/2023]
Abstract
The epidemic of obesity and its co-mortalities has reached an alarming level worldwide. Currently, metabolic surgeries, especially the Roux-en-Y gastric bypass and vertical sleeve gastrectomy, are the most effective and sustainable treatments for obesity, type 2 diabetes, non-alcoholic steatohepatitis, as well as other metabolic diseases. However, the invasive nature of the surgeries limits their broad applications to the general public. Therefore, developing alternative non-invasive approaches to mimic metabolic surgery is an important direction of the field. Recent studies have identified several potential metabolic surgery-induced downstream endocrine mediators, among which bile acids are key candidate signaling molecules. Bile acids are profoundly altered by metabolic surgery, which contributes to the metabolic effects of the surgery. In this review, we focus on the most recent studies on the roles of bile acids and bile acid receptors farnesoid X receptor and Takeda G protein-coupled receptor 5 in mediating the metabolic effects of metabolic surgery. We conclude that targeting bile acid pathways may be a promising pharmacological approach to mimic the beneficial effects of metabolic surgery.
Collapse
Affiliation(s)
- Hui Xue
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Luyao Huang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jui Tu
- Department of Diabetes Complications and Metabolism, Institute of Diabetes and Metabolism Research Center, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Lili Ding
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Institute of Diabetes and Metabolism Research Center, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
26
|
Effect of Gegen Qinlian Decoction on Hepatic Gluconeogenesis in ZDF Rats with Type 2 Diabetes Mellitus Based on the Farnesol X Receptor/Ceramide Signaling Pathway Regulating Mitochondrial Metabolism and Endoplasmic Reticulum Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9922292. [PMID: 34422083 PMCID: PMC8371656 DOI: 10.1155/2021/9922292] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022]
Abstract
Background Type 2 diabetes mellitus (T2DM) is a kind of disorder of glucose and lipid metabolism with the main clinical manifestation of long‐term higher blood glucose level than the normal value. Farnesol X receptor (FXR)/ceramide signaling pathway plays an important role in regulating cholesterol metabolism, lipid homeostasis, and the absorption of fat and vitamins in diet. Gegen Qinlian Decoction (GQD) is a classical herbal formula, which has a good clinical therapeutic effect on diabetes-related metabolic syndrome. Objective To investigate the effect of Gegen Qinlian Decoction (GQD) on hepatic gluconeogenesis in obese T2DM rats based on the FXR/ceramide signaling pathway regulating mitochondrial metabolism and endoplasmic reticulum stress (ERS). Methods ZDF (fa/fa) rats were fed with high-fat diet to establish the T2DM model; GQD was given to T2DM model rats by gavage; changes of the general state and body weight of rats were recorded; fasting blood glucose was detected; blood insulin, blood ceramide, glycosylated hemoglobin in blood, acetyl CoA in liver mitochondria, and bile salt lyase in intestinal tissue were detected by ELISA. The content of T-β-MCA in blood was detected by LC-MS; the content of glycogen in liver tissue was detected by PAS staining; the expression of FXR, Sptlc2, and Smpd3 in ileum tissue, P-PERK, ATF6α, GRP78 BIP, and P-IRE1 in the liver, and CS and PC protein in liver mitochondria was detected by immunohistochemistry and western blot assay. The mRNA expression levels of FXR, Sptlc2, and Smpd3 in the ileum, PERK, ATF6α, GRP78 BIP, and IRE1 in the liver, and CS and PC in liver mitochondria were detected by qRT-PCR. Results GQD can improve the general state of T2DM rats, slow down their weight gain, reduce the levels of fasting blood glucose, fasting insulin, glycosylated hemoglobin, blood ceramide, bile salt hydrolase in intestinal tissue, and acetyl CoA in liver mitochondria of T2DM rats, and increase the contents of liver glycogen and T-β-MCA in blood of T2DM rats. At the molecular level, GQD can inhibit the expression levels of FXR, Sptlc2, and Smpd3 in the ileum of T2DM rats and the protein and mRNA expression levels of oxidative stress-related factors in the liver. At the same time, GQD can increase the expression of CS and reduce the expression of PC in liver mitochondria of T2DM rats. Conclusion GQD can inhibit the FXR/ceramide signaling pathway, regulate endoplasmic reticulum stress, enhance the CS activity of liver mitochondria, reduce the acetyl CoA level and PC activity of liver mitochondria, inhibit hepatic gluconeogenesis, protect islet β-cells, and control blood glucose.
Collapse
|
27
|
Tu Y, Bao Y, Zhang P. Metabolic surgery in China: present and future. J Mol Cell Biol 2021; 13:mjab039. [PMID: 34240190 PMCID: PMC8697345 DOI: 10.1093/jmcb/mjab039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/27/2021] [Accepted: 06/14/2021] [Indexed: 11/24/2022] Open
Abstract
Obesity and its related complications comprise a serious public health problem worldwide, and obesity is increasing in China. Metabolic surgery is a new type of treatment with unique advantages in weight loss and obesity-related metabolic complications. The pathogenesis of obesity is complex and not yet fully understood. Here, we review the current efficacy and safety of metabolic surgery, as well as recent progress in mechanistic studies and surgical procedures in China. The exciting and rapid advances in this field provide new opportunities for patients with obesity and strike a balance between long-term effectiveness and safety.
Collapse
Affiliation(s)
- Yinfang Tu
- Department of Endocrinology and Metabolism, Shanghai
Jiao Tong University Affiliated Sixth People’s HospitalShanghai Diabetes
Institute, Shanghai Clinical Center of Diabetes, Shanghai Key Laboratory of
Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic
Disease, Shanghai 200233, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai
Jiao Tong University Affiliated Sixth People’s HospitalShanghai Diabetes
Institute, Shanghai Clinical Center of Diabetes, Shanghai Key Laboratory of
Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic
Disease, Shanghai 200233, China
- Department of Endocrinology and Metabolism, Jinshan
District Central Hospital of Shanghai Sixth People's
Hospital, Shanghai 201599, China
| | - Pin Zhang
- Department of Bariatric and Metabolic Surgery,
Shanghai Jiao Tong University Affiliated Sixth People’s
Hospital, Shanghai 200233, China
| |
Collapse
|
28
|
Bousquet J, Czarlewski W, Zuberbier T, Mullol J, Blain H, Cristol JP, De La Torre R, Pizarro Lozano N, Le Moing V, Bedbrook A, Agache I, Akdis CA, Canonica GW, Cruz AA, Fiocchi A, Fonseca JA, Fonseca S, Gemicioğlu B, Haahtela T, Iaccarino G, Ivancevich JC, Jutel M, Klimek L, Kraxner H, Kuna P, Larenas-Linnemann DE, Martineau A, Melén E, Okamoto Y, Papadopoulos NG, Pfaar O, Regateiro FS, Reynes J, Rolland Y, Rouadi PW, Samolinski B, Sheikh A, Toppila-Salmi S, Valiulis A, Choi HJ, Kim HJ, Anto JM. Potential Interplay between Nrf2, TRPA1, and TRPV1 in Nutrients for the Control of COVID-19. Int Arch Allergy Immunol 2021; 182:324-338. [PMID: 33567446 PMCID: PMC8018185 DOI: 10.1159/000514204] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022] Open
Abstract
In this article, we propose that differences in COVID-19 morbidity may be associated with transient receptor potential ankyrin 1 (TRPA1) and/or transient receptor potential vanilloid 1 (TRPV1) activation as well as desensitization. TRPA1 and TRPV1 induce inflammation and play a key role in the physiology of almost all organs. They may augment sensory or vagal nerve discharges to evoke pain and several symptoms of COVID-19, including cough, nasal obstruction, vomiting, diarrhea, and, at least partly, sudden and severe loss of smell and taste. TRPA1 can be activated by reactive oxygen species and may therefore be up-regulated in COVID-19. TRPA1 and TRPV1 channels can be activated by pungent compounds including many nuclear factor (erythroid-derived 2) (Nrf2)-interacting foods leading to channel desensitization. Interactions between Nrf2-associated nutrients and TRPA1/TRPV1 may be partly responsible for the severity of some of the COVID-19 symptoms. The regulation by Nrf2 of TRPA1/TRPV1 is still unclear, but suggested from very limited clinical evidence. In COVID-19, it is proposed that rapid desensitization of TRAP1/TRPV1 by some ingredients in foods could reduce symptom severity and provide new therapeutic strategies.
Collapse
Affiliation(s)
- Jean Bousquet
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Charité, and Berlin Institute of Health, Comprehensive Allergy Center, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany, .,University Hospital and MACVIA France, Montpellier, France,
| | | | - Torsten Zuberbier
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Charité, and Berlin Institute of Health, Comprehensive Allergy Center, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Joaquim Mullol
- Rhinology Unit & Smell Clinic, ENT Department, Hospital Clinic - Clinical & Experimental Respiratory Immunoallergy, IDIBAPS, CIBERES, Universitat de Barcelona, Barcelona, Spain
| | - Hubert Blain
- Department of Geriatrics, Montpellier University Hospital, Montpellier, France
| | - Jean-Paul Cristol
- Laboratoire de Biochimie et Hormonologie, PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de, Montpellier, France
| | - Rafael De La Torre
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.,IMIM (Hospital del Mar Research Institute), Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut Toxicologia, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | | | - Anna Bedbrook
- University Hospital and MACVIA France, Montpellier, France.,MASK-air, Montpellier, France
| | - Ioana Agache
- Faculty of Medicine, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - G Walter Canonica
- Personalized Medicine, Asthma and Allergy, Humanitas Clinical and Research Center IRCCS and Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Alvaro A Cruz
- Fundação ProAR, Federal University of Bahia and GARD/WHO Planning Group, Salvador, Brazil
| | - Alessandro Fiocchi
- Division of Allergy, The Bambino Gesù Children's Research Hospital IRCCS, Rome, Italy
| | - Joao A Fonseca
- CINTESIS, Center for Research in Health Technologies and Information Systems, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,MEDIDA, Lda, Porto, Portugal
| | - Susana Fonseca
- GreenUPorto - Sustainable Agrifood Production Research Centre, DGAOT, Faculty of Sciences, University of Porto, Vila do Conde, Portugal
| | - Bilun Gemicioğlu
- Department of Pulmonary Diseases, Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital, and University of Helsinki, Helsinki, Finland
| | - Guido Iaccarino
- Interdepartmental Center of Research on Hypertension and Related Conditions CIRIAPA, Federico II University, Napoli, Italy
| | | | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University and ALL-MED Medical Research Institute, Wrocław, Poland
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Helga Kraxner
- Department of Otorhinolaryngology, Head and Neck Surgery, Semmelweis University, Budapest, Hungary
| | - Piotr Kuna
- Division of Internal Medicine, Asthma and Allergy, Barlicki University Hospital, Medical University of Lodz, Lodz, Poland
| | - Désirée E Larenas-Linnemann
- Center of Excellence in Asthma and Allergy, Médica Sur Clinical Foundation and Hospital, Mexico City, Mexico
| | - Adrian Martineau
- Institute for Population Health Sciences, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Sachs' Children's Hospital, Stockholm, Sweden
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology, Chiba University Hospital, Chiba, Japan
| | - Nikolaos G Papadopoulos
- Division of Infection, Immunity & Respiratory Medicine, Royal Manchester Children's Hospital, University of Manchester, Manchester, United Kingdom.,Allergy Department, 2nd Pediatric Clinic, Athens General Children's Hospital "P&A Kyriakou," University of Athens, Athens, Greece
| | - Oliver Pfaar
- Section of Rhinology and Allergy, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Frederico S Regateiro
- Allergy and Clinical Immunology Unit, Centro Hospitalar e Universitário de Coimbra, Faculty of Medicine, Institute of Immunology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, ICBR - Institute for Clinical and Biomedical Research, CIBB, University of Coimbra, Coimbra, Portugal
| | - Jacques Reynes
- Maladies Infectieuses et Tropicales, CHU, Montpellier, France
| | | | - Philip W Rouadi
- Department of Otolaryngology-Head and Neck Surgery, Eye and Ear University Hospital, Beirut, Lebanon
| | - Boleslaw Samolinski
- Department of Prevention of Environmental Hazards and Allergology, Medical University of Warsaw, Warsaw, Poland
| | - Aziz Sheikh
- Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Sanna Toppila-Salmi
- Skin and Allergy Hospital, Helsinki University Hospital, and University of Helsinki, Helsinki, Finland
| | - Arunas Valiulis
- Vilnius University Faculty of Medicine, Institute of Clinical Medicine & Institute of Health Sciences, Vilnius, Lithuania
| | - Hak-Jong Choi
- Research and Development Division, Microbiology and Functionality Research Group, World Institute of Kimchi, Gwangju, Republic of Korea
| | - Hyun Ju Kim
- Strategy and Planning Division, SME Service Department, World Institute of Kimchi, Gwangju, Republic of Korea
| | - Josep M Anto
- IMIM (Hospital del Mar Research Institute), Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut Toxicologia, Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.,ISGlobAL, Barcelona, Centre for Research in Environmental Epidemiology, Barcelona, Spain
| |
Collapse
|
29
|
Miskelly MG, Shcherbina L, Thorén Fischer AH, Abels M, Lindqvist A, Wierup N. GK-rats respond to gastric bypass surgery with improved glycemia despite unaffected insulin secretion and beta cell mass. Peptides 2021; 136:170445. [PMID: 33197511 DOI: 10.1016/j.peptides.2020.170445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Roux-en-Y gastric bypass (RYGB) is the most effective treatment for morbid obesity and results in rapid remission of type 2 diabetes (T2D), before significant weight loss occurs. The underlying mechanisms for T2D remission are not fully understood. To gain insight into these mechanisms we used RYGB-operated diabetic GK-rats and Wistar control rats. Twelve adult male Wistar- and twelve adult male GK-rats were subjected to RYGB- or sham-operation. Oral glucose tolerance tests (OGTT) were performed six weeks after surgery. RYGB normalized fasting glucose levels in GK-rats, without affecting fasting insulin levels. In both rat strains, RYGB caused increased postprandial responses in glucose, GLP-1, and GIP. RYGB caused elevated postprandial insulin secretion in Wistar-rats, but had no effect on insulin secretion in GK-rats. In agreement with this, RYGB improved HOMA-IR in GK-rats, but had no effect on HOMA-β. RYGB-operated GK-rats had an increased number of GIP receptor and GLP-1 receptor immunoreactive islet cells, but RYGB had no major effect on beta or alpha cell mass. Furthermore, in RYGB-operated GK-rats, increased Slc5a1, Pck2 and Pfkfb1 and reduced Fasn hepatic mRNA expression was observed. In summary, our data shows that RYGB induces T2D remission and enhanced postprandial incretin hormone secretion in GK-rats, without affecting insulin secretion or beta cell mass. Thus our data question the dogmatic view of how T2D remission is achieved and instead point at improved insulin sensitivity as the main mechanism of remission.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/surgery
- Disease Models, Animal
- Gastric Bypass
- Gastric Inhibitory Polypeptide/genetics
- Glucagon-Like Peptide 1/genetics
- Glucose Tolerance Test
- Humans
- Insulin/genetics
- Insulin/metabolism
- Insulin Secretion/genetics
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Obesity, Morbid/genetics
- Obesity, Morbid/metabolism
- Obesity, Morbid/pathology
- Obesity, Morbid/surgery
- Rats
- Rats, Wistar
- Weight Loss/genetics
- Weight Loss/physiology
Collapse
Affiliation(s)
- Michael G Miskelly
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | - Liliya Shcherbina
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | | | - Mia Abels
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | - Andreas Lindqvist
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | - Nils Wierup
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden.
| |
Collapse
|
30
|
Islam MS. Molecular Regulations and Functions of the Transient Receptor Potential Channels of the Islets of Langerhans and Insulinoma Cells. Cells 2020; 9:cells9030685. [PMID: 32168890 PMCID: PMC7140661 DOI: 10.3390/cells9030685] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 12/17/2022] Open
Abstract
Insulin secretion from the β-cells of the islets of Langerhans is triggered mainly by nutrients such as glucose, and incretin hormones such as glucagon-like peptide-1 (GLP-1). The mechanisms of the stimulus-secretion coupling involve the participation of the key enzymes that metabolize the nutrients, and numerous ion channels that mediate the electrical activity. Several members of the transient receptor potential (TRP) channels participate in the processes that mediate the electrical activities and Ca2+ oscillations in these cells. Human β-cells express TRPC1, TRPM2, TRPM3, TRPM4, TRPM7, TRPP1, TRPML1, and TRPML3 channels. Some of these channels have been reported to mediate background depolarizing currents, store-operated Ca2+ entry (SOCE), electrical activity, Ca2+ oscillations, gene transcription, cell-death, and insulin secretion in response to stimulation by glucose and GLP1. Different channels of the TRP family are regulated by one or more of the following mechanisms: activation of G protein-coupled receptors, the filling state of the endoplasmic reticulum Ca2+ store, heat, oxidative stress, or some second messengers. This review briefly compiles our current knowledge about the molecular mechanisms of regulations, and functions of the TRP channels in the β-cells, the α-cells, and some insulinoma cell lines.
Collapse
Affiliation(s)
- Md. Shahidul Islam
- Karolinska Institutet, Department of Clinical Science and Education, Södersjukhuset, Research Center, 5th floor, SE-118 83 Stockholm, Sweden;
- Department of Emergency Care and Internal Medicine, Uppsala University Hospital, Uppsala University, SE-751 85 Uppsala, Sweden
| |
Collapse
|