1
|
Oshiro A, Aotani R, Sakamoto W, Kitazono T, Ohkuma T. Long-Term Effects of Incretin-Based Drugs on Glycemic Control in Permanent Neonatal Diabetes. JCEM CASE REPORTS 2024; 2:luae188. [PMID: 39430732 PMCID: PMC11487289 DOI: 10.1210/jcemcr/luae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Indexed: 10/22/2024]
Abstract
Permanent neonatal diabetes mellitus (PNDM) is a genetic disorder, characterized by a decrease in endogenous insulin secretion. Therefore, exogenous insulin supplementation plays a central role in controlling glycemia. Although adding a sulfonylurea can help to discontinue insulin, discontinuation is sometimes difficult when the sulfonylurea is administered at older ages. A 24-year-old woman with longstanding PNDM who had poor glycemic control using insulin (47 U/day) and high-dose glibenclamide (0.6 mg/kg/day), had successfully discontinued insulin after initiating the dipeptidyl peptidase-4 inhibitor sitagliptin (50 mg/day). Additionally, hemoglobin A1c levels decreased by 4.8%. Double dosing of sitagliptin and subsequent switching to the glucagon-like peptide-1 receptor agonist semaglutide (0.25 mg/week followed by 0.5 mg/week) further decreased hemoglobin A1c values, with graded improvements in endogenous insulin secretion. There were no episodes of hypoglycemia during which glibenclamide was titrated down from 0.6 to 0.4 mg/kg/day. Intra- and inter-day glucose variability as assessed by continuous glucose monitoring was also improved. In patients with PNDM, administration and dose escalation of incretin-based drugs, in addition to a high-dose sulfonylurea, could be a useful treatment strategy. This strategy may be helpful for discontinuing insulin, downtitrating sulfonylureas, and subsequent achievement of better glycemic control regarding long-term stability and short-term variability.
Collapse
Affiliation(s)
- Ayaka Oshiro
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryoichiro Aotani
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Wakako Sakamoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Toshiaki Ohkuma
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
2
|
Gonzalez CE, Vaidya RS, Clayton SW, Tang SY. Secreted chemokines reveal diverse inflammatory and degenerative processes in the intervertebral disc of the STZ-HFD mouse model of Type 2 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605332. [PMID: 39131361 PMCID: PMC11312574 DOI: 10.1101/2024.07.31.605332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The chronic inflammation present in type 2 diabetes causes many chronic inflammatory comorbidities, including cardiovascular, renal, and neuropathic complications. Type 2 diabetes is also associated with a number of spinal pathologies, including intervertebral disc (IVD) degeneration and chronic neck and back pain. Although confounding factors such as obesity are thought to increase the loads to the musculoskeletal system and subsequent degeneration, studies have shown that even after adjusting age, body mass index, and genetics (e.g. twins), patients with diabetes suffer from disproportionately more IVD degeneration and back pain. Yet the tissue-specific responses of the IVD during diabetes remains relatively unknown. We hypothesize that chronic diabetes fosters a proinflammatory microenvironment within the IVD that accelerates degeneration and increases susceptibility to painful disorders. To test this hypothesis, we evaluated two commonly used mouse models of diabetes - the leptin-receptor deficient mouse (db/db) and the chronic high-fat diet in mice with impaired beta-cell function (STZ-HFD). The db/db is a genetic model that spontaneous develop diabetes through hyperphagia, while the STZ-HFD mouse first exhibits rapid obesity development under HFD and pronounced insulin resistance following streptozotocin administration. Both animal models were allowed to develop sustained diabetes for at least twelve weeks, as defined by elevated hemoglobin A1C, hyperglycemia, and glucose intolerance. Following the twelve-week period, the IVDs were extracted in quantified in several measures including tissue-specific secreted cytokines, viscoelastic mechanical behavior, structural composition, and histopathologic degeneration. Although there were no differences in mechanical function or the overall structure of the IVD, the STZ-HFD IVDs were more degenerated. More notably, the STZ-HFD model shows a significantly higher fold increase for eight cytokines: CXCL2, CCL2, CCL3, CCL4, CCL12 (monocyte/macrophage associated), IL-2, CXCL9 (T-cell associated), and CCL5 (pleiotropic). Correlative network analyses revealed that the expression of cytokines differentially regulated between the db/db and the STZ-HFD models. Moreover, the STZ-HFD contained a fragmented and modular cytokine network, indicating greater complexities in the regulatory network. Taken together, the STZ-HFD model of type 2 diabetes may better recapitulate the complexities of the chronic inflammatory processes in the IVD during diabetes.
Collapse
Affiliation(s)
- Christian E. Gonzalez
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
| | - Rachana S. Vaidya
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| | - Sade W. Clayton
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| | - Simon Y. Tang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
- Institute of Material Science and Engineering, Washington University in St. Louis, St. Louis, MO
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
3
|
Moon JH, Choe HJ, Lim S. Pancreatic beta-cell mass and function and therapeutic implications of using antidiabetic medications in type 2 diabetes. J Diabetes Investig 2024; 15:669-683. [PMID: 38676410 PMCID: PMC11143426 DOI: 10.1111/jdi.14221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/23/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Nowadays, the focus of diabetes treatment has switched from lowering the glucose level to preserving glycemic homeostasis and slowing the disease progression. The main pathophysiology of both type 1 diabetes and long-standing type 2 diabetes is pancreatic β-cell mass loss and dysfunction. According to recent research, human pancreatic β-cells possess the ability to proliferate in response to elevated insulin demands. It has been demonstrated that in insulin-resistant conditions in humans, such as obesity or pregnancy, the β-cell mass increases. This ability could be helpful in developing novel treatment approaches to restore a functional β-cell mass. Treatment strategies aimed at boosting β-cell function and mass may be a useful tool for managing diabetes mellitus and stopping its progression. This review outlines the processes of β-cell failure and detail the many β-cell abnormalities that manifest in people with diabetes mellitus. We also go over standard techniques for determining the mass and function of β-cells. Lastly, we provide the therapeutic implications of utilizing antidiabetic drugs in controlling the mass and function of pancreatic β-cells.
Collapse
Affiliation(s)
- Joon Ho Moon
- Department of Internal MedicineSeoul National University College of MedicineSeongnamSouth Korea
- Department of Internal MedicineSeoul National University Bundang HospitalSeongnamSouth Korea
| | - Hun Jee Choe
- Department of Internal MedicineHallym University Dongtan Sacred Heart HospitalHwaseongSouth Korea
| | - Soo Lim
- Department of Internal MedicineSeoul National University College of MedicineSeongnamSouth Korea
- Department of Internal MedicineSeoul National University Bundang HospitalSeongnamSouth Korea
| |
Collapse
|
4
|
Bouchareb EM, Derbal K, Bedri R, Slimani K, Menas S, Lazreg H, Maaref F, Ouabdelkader S, Saheb A, Bouaita R, Bouchareb R, Dizge N. Improving Biohydrogen Production by Dark Fermentation of Milk Processing Wastewater by Physicochemical and Enzymatic Pretreatments. Appl Biochem Biotechnol 2024; 196:2741-2756. [PMID: 37682509 DOI: 10.1007/s12010-023-04619-2] [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] [Accepted: 07/01/2023] [Indexed: 09/09/2023]
Abstract
Biohydrogen is considered an alternative energy reserve. Dark fermentation is one of the important green hydrogen production techniques that utilizes organic waste as raw material. It is a promising bioconversion, easy, not expensive, and cost-effective process. Milk processing wastewater (MPWW) is an organic effluent generated in large volumes on a daily basis and disposed directly into the environment. In this research, the study of biochemical hydrogen potential (BHP) test of MPWW was evaluated and used as substrate (S). A waste sludge was used as an inoculum (I) and source of bacteria. Both substrate and inoculum were analyzed and the study was based mainly on the ratio of volatile solids (VS) of inoculum and substrate subsequently, which was noted as I/S. Different substrate pretreatments were performed: ultrasonic, thermal, chemical, and enzymatic hydrolysis. The I/S ratio impact was investigated and evaluated the hydrogen production improvement. Modified Gompertz and modified Logistic kinetic models were employed for the kinetic modeling of cumulative hydrogen production values. Results show that I/S ratio of 1/4 gVS/gVS resulted from the best hydrogen production of 59.96 mL during 30 days of MPWW fermentation without pretreatment. It was also shown that all the adopted pretreatments enhanced hydrogen production, whereas ultrasonic pretreatment for 5 min increased the production by only 14.84%. Heat pretreatment was more efficient, where the hydrogen production increased from 60 to 162 mL (170% of improvement) using heat shock at 90 °C for 30 min. The impact of chemical pretreatment was different from a reagent to another. Pretreatment using calcium hydroxide resulted in the biggest hydrogen production of 165.3 mL (175.5%) compared to the other chemical pretreatments. However, the best hydrogen production was given by the biological pretreatment using enzymatic hydrolysis (Lactase) resulting in 254 mL of hydrogen production, which is equivalent to 323.62% of production improvement. Modified Gompertz and Logistic kinetic models fitted well with experimental data. Thus, the enzymatic hydrolysis of MPWW proved to be a promising technique for biohydrogen production enhancement.
Collapse
Affiliation(s)
- Esma Mahfouf Bouchareb
- Department of Environmental Engineering, Process Engineering Faculty, Saleh Boubnider University, 25000, Constantine, Algeria
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
- Laboratory of Process Engineering for Sustainable Development and Health Products (LGPDDPS), National Polytechnic School of Constantine, 25000, Constantine, Algeria
| | - Kerroum Derbal
- Laboratory of Process Engineering for Sustainable Development and Health Products (LGPDDPS), National Polytechnic School of Constantine, 25000, Constantine, Algeria
- Department of Process Engineering, National High School of Polytechnic, Malek Bennabi, Constantine 3, Algeria
| | - Rayane Bedri
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
| | - Khaled Slimani
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
| | - Souha Menas
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
| | - Halima Lazreg
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
| | - Feriel Maaref
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
| | - Samir Ouabdelkader
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
| | - Aya Saheb
- Department of Engineering, National High School of Biotechnology, Toufik Khaznadar, Constantine 3, Algeria
| | - Rokaya Bouaita
- Department of Process Engineering, National High School of Polytechnic, Malek Bennabi, Constantine 3, Algeria
| | - Raouf Bouchareb
- Department of Environmental Engineering, Process Engineering Faculty, Saleh Boubnider University, 25000, Constantine, Algeria
- Laboratory of Process Engineering for Sustainable Development and Health Products (LGPDDPS), National Polytechnic School of Constantine, 25000, Constantine, Algeria
| | - Nadir Dizge
- Department of Environmental Engineering, Mersin University, 33343, Mersin, Turkey.
| |
Collapse
|
5
|
Bosch AJT, Rohm TV, AlAsfoor S, Low AJY, Keller L, Baumann Z, Parayil N, Stawiski M, Rachid L, Dervos T, Mitrovic S, Meier DT, Cavelti-Weder C. Lung versus gut exposure to air pollution particles differentially affect metabolic health in mice. Part Fibre Toxicol 2023; 20:7. [PMID: 36895000 PMCID: PMC9996885 DOI: 10.1186/s12989-023-00518-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Air pollution has emerged as an unexpected risk factor for diabetes. However, the mechanism behind remains ill-defined. So far, the lung has been considered as the main target organ of air pollution. In contrast, the gut has received little scientific attention. Since air pollution particles can reach the gut after mucociliary clearance from the lungs and through contaminated food, our aim was to assess whether exposure deposition of air pollution particles in the lung or the gut drive metabolic dysfunction in mice. METHODS To study the effects of gut versus lung exposure, we exposed mice on standard diet to diesel exhaust particles (DEP; NIST 1650b), particulate matter (PM; NIST 1649b) or phosphate-buffered saline by either intratracheal instillation (30 µg 2 days/week) or gavage (12 µg 5 days/week) over at least 3 months (total dose of 60 µg/week for both administration routes, equivalent to a daily inhalation exposure in humans of 160 µg/m3 PM2.5) and monitored metabolic parameters and tissue changes. Additionally, we tested the impact of the exposure route in a "prestressed" condition (high-fat diet (HFD) and streptozotocin (STZ)). RESULTS Mice on standard diet exposed to particulate air pollutants by intratracheal instillation developed lung inflammation. While both lung and gut exposure resulted in increased liver lipids, glucose intolerance and impaired insulin secretion was only observed in mice exposed to particles by gavage. Gavage with DEP created an inflammatory milieu in the gut as shown by up-regulated gene expression of pro-inflammatory cytokines and monocyte/macrophage markers. In contrast, liver and adipose inflammation markers were not increased. Beta-cell secretory capacity was impaired on a functional level, most likely induced by the inflammatory milieu in the gut, and not due to beta-cell loss. The differential metabolic effects of lung and gut exposures were confirmed in a "prestressed" HFD/STZ model. CONCLUSIONS We conclude that separate lung and gut exposures to air pollution particles lead to distinct metabolic outcomes in mice. Both exposure routes elevate liver lipids, while gut exposure to particulate air pollutants specifically impairs beta-cell secretory capacity, potentially instigated by an inflammatory milieu in the gut.
Collapse
Affiliation(s)
- Angela J T Bosch
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Theresa V Rohm
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Shefaa AlAsfoor
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Andy J Y Low
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Lena Keller
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Zora Baumann
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Neena Parayil
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Marc Stawiski
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Leila Rachid
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Thomas Dervos
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Sandra Mitrovic
- Department of Laboratory Medicine, University Hospital Basel, 4031, Basel, Switzerland
| | - Daniel T Meier
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Claudia Cavelti-Weder
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland. .,Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland. .,Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Rämistrasse 100, 8009, Zurich, Switzerland.
| |
Collapse
|
6
|
Zhang Y, Han S, Liu C, Zheng Y, Li H, Gao F, Bian Y, Liu X, Liu H, Hu S, Li Y, Chen ZJ, Zhao S, Zhao H. THADA inhibition in mice protects against type 2 diabetes mellitus by improving pancreatic β-cell function and preserving β-cell mass. Nat Commun 2023; 14:1020. [PMID: 36823211 PMCID: PMC9950491 DOI: 10.1038/s41467-023-36680-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Impaired insulin secretion is a hallmark in type 2 diabetes mellitus (T2DM). THADA has been identified as a candidate gene for T2DM, but its role in glucose homeostasis remains elusive. Here we report that THADA is strongly activated in human and mouse islets of T2DM. Both global and β-cell-specific Thada-knockout mice exhibit improved glycemic control owing to enhanced β-cell function and decreased β-cell apoptosis. THADA reduces endoplasmic reticulum (ER) Ca2+ stores in β-cells by inhibiting Ca2+ re-uptake via SERCA2 and inducing Ca2+ leakage through RyR2. Upon persistent ER stress, THADA interacts with and activates the pro-apoptotic complex comprising DR5, FADD and caspase-8, thus aggravating ER stress-induced apoptosis. Importantly, THADA deficiency protects mice from high-fat high-sucrose diet- and streptozotocin-induced hyperglycemia by restoring insulin secretion and preserving β-cell mass. Moreover, treatment with alnustone inhibits THADA's function, resulting in ameliorated hyperglycemia in obese mice. Collectively, our results support pursuit of THADA as a potential target for developing T2DM therapies.
Collapse
Affiliation(s)
- Yuqing Zhang
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Shan Han
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Congcong Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Yuanwen Zheng
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong, China
| | - Hao Li
- Shandong Provincial Qianfoshan Hospital, Shandong University, 250014, Jinan, Shandong, China
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Science, 100101, Beijing, China
| | - Yuehong Bian
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Xin Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Shourui Hu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Yuxuan Li
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 200135, Shanghai, China. .,Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, 250012, Jinan, China.
| | - Shigang Zhao
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
| |
Collapse
|
7
|
Wu X, Liu H, Brooks A, Xu S, Luo J, Steiner R, Mickelsen DM, Moravec CS, Jeffrey AD, Small EM, Jin ZG. SIRT6 Mitigates Heart Failure With Preserved Ejection Fraction in Diabetes. Circ Res 2022; 131:926-943. [PMID: 36278398 PMCID: PMC9669223 DOI: 10.1161/circresaha.121.318988] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 10/13/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a growing health problem without effective therapies. Epidemiological studies indicate that diabetes is a strong risk factor for HFpEF, and about 45% of patients with HFpEF are suffering from diabetes, yet the underlying mechanisms remain elusive. METHODS Using a combination of echocardiography, hemodynamics, RNA-sequencing, molecular biology, in vitro and in vivo approaches, we investigated the roles of SIRT6 (sirtuin 6) in regulation of endothelial fatty acid (FA) transport and HFpEF in diabetes. RESULTS We first observed that endothelial SIRT6 expression was markedly diminished in cardiac tissues from heart failure patients with diabetes. We then established an experimental mouse model of HFpEF in diabetes induced by a combination of the long-term high-fat diet feeding and a low-dose streptozocin challenge. We also generated a unique humanized SIRT6 transgenic mouse model, in which a single copy of human SIRT6 transgene was engineered at mouse Rosa26 locus and conditionally induced with the Cre-loxP technology. We found that genetically restoring endothelial SIRT6 expression in the diabetic mice ameliorated diastolic dysfunction concurrently with decreased cardiac lipid accumulation. SIRT6 gain- or loss-of-function studies showed that SIRT6 downregulated endothelial FA uptake. Mechanistically, SIRT6 suppressed endothelial expression of PPARγ through SIRT6-dependent deacetylation of histone H3 lysine 9 around PPARγ promoter region; and PPARγ reduction mediated SIRT6-dependent inhibition of endothelial FA uptake. Importantly, oral administration of small molecule SIRT6 activator MDL-800 to diabetic mice mitigated cardiac lipid accumulation and diastolic dysfunction. CONCLUSIONS The impairment of endothelial SIRT6 expression links diabetes to HFpEF through the alteration of FA transport across the endothelial barrier. Genetic and pharmacological strategies that restored endothelial SIRT6 function in mice with diabetes alleviated experimental HFpEF by limiting FA uptake and improving cardiac metabolism, thus warranting further clinical evaluation.
Collapse
Affiliation(s)
- Xiaoqian Wu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Alan Brooks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jinque Luo
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Rebbeca Steiner
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Deanne M. Mickelsen
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christine S. Moravec
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Alexis D. Jeffrey
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Eric M. Small
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
8
|
Gasparini P, Garofolo IC, Telles MM, Oyama LM, Veneza VDM, Moura Veiga TA, Flor Silveira VL, Caperuto LC. Bauhinia forficata link extract attenuates insulin resistance by preserving glucose uptake in gastrocnemius muscle. Nat Prod Res 2022; 37:2031-2036. [PMID: 35997243 DOI: 10.1080/14786419.2022.2113875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Bioactive metabolites from Bauhinia forficata Link (Bf extract) hold therapeutic potential for type 2 diabetes mellitus (T2DM) but the mechanism remains poorly understood. This study aimed to test the extract from Bf leaves obtained by decoction on the prevention of T2DM in vivo. The Bf extract was tested on a streptozotocin-induced T2DM mouse model fed on a high-fat diet. The insulin resistance was attenuated in T2DM animals supplemented with Bf extract, which indicates glucose intolerance reduction and p-AKT/AKT ratio preservation in the gastrocnemius muscle. These observations suggested that Bf extract enhanced glucose uptake. Nevertheless, there was no preservation in β-cell insulin secretion in Bf extract-treated T2DM mice. Interestingly, the Bf extract reduced body weight gain without affecting total energy intake. Hence, Bf extract has a hypoglycemic effect which could attenuate the development of insulin resistance.
Collapse
Affiliation(s)
- Patricia Gasparini
- Departamento de Ciências Biológicas; Instituto de Ciências Ambientais, Químicas e Farmacêuticas (ICAQF); Universidade Federal de São Paulo; Diadema, Brazil
| | - Ingrid Candido Garofolo
- Departamento de Ciências Biológicas; Instituto de Ciências Ambientais, Químicas e Farmacêuticas (ICAQF); Universidade Federal de São Paulo; Diadema, Brazil
| | - Monica Marques Telles
- Departamento de Ciências Biológicas; Instituto de Ciências Ambientais, Químicas e Farmacêuticas (ICAQF); Universidade Federal de São Paulo; Diadema, Brazil
| | - Lila Missae Oyama
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Viviane de Mello Veneza
- Departamento de Química; Instituto de Ciências Ambientais, Químicas e Farmacêuticas (ICAQF); Universidade Federal de São Paulo, Diadema, Brazil
| | - Thiago André Moura Veiga
- Departamento de Química; Instituto de Ciências Ambientais, Químicas e Farmacêuticas (ICAQF); Universidade Federal de São Paulo, Diadema, Brazil
| | - Vera Lucia Flor Silveira
- Departamento de Ciências Biológicas; Instituto de Ciências Ambientais, Químicas e Farmacêuticas (ICAQF); Universidade Federal de São Paulo; Diadema, Brazil
| | - Luciana Chagas Caperuto
- Departamento de Ciências Biológicas; Instituto de Ciências Ambientais, Químicas e Farmacêuticas (ICAQF); Universidade Federal de São Paulo; Diadema, Brazil
| |
Collapse
|
9
|
Therapeutic Perspectives of CD26 Inhibitors in Imune-Mediated Diseases. Molecules 2022; 27:molecules27144498. [PMID: 35889373 PMCID: PMC9321265 DOI: 10.3390/molecules27144498] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/05/2022] [Accepted: 07/10/2022] [Indexed: 02/01/2023] Open
Abstract
The enzymatic activity of CD26/DPP4 (dipeptidyl peptidase 4/DPP4) is highlighted in multiple studies to play a vital role in glucose metabolism by cleaving and inactivating the incretins glucagon-like peptide-1 (GLP) and gastric inhibitory protein (GIP). A large number of studies demonstrate that CD26 also plays an integral role in the immune system, particularly in T cell activation. CD26 is extensively expressed in immune cells, such as T cells, B cells, NK cells, dendritic cells, and macrophages. The enzymatic activity of CD26 cleaves and regulates numerous chomokines and cytokines. CD26 inhibitors have been widely used for the treatment of diabetes mellitus, while it is still under investigation as a therapy for immune-mediated diseases. In addition, CD26’s involvement in cancer immunology was also described. The review aims to summarize the therapeutic effects of CD26 inhibitors on immune-mediated diseases, as well as the mechanisms that underpin them.
Collapse
|
10
|
Esser N, Schmidt C, Barrow BM, Cronic L, Hackney DJ, Mongovin SM, Hogan MF, Templin AT, Castillo JJ, Hull RL, Zraika S. Insulinotropic Effects of Neprilysin and/or Angiotensin Receptor Inhibition in Mice. Front Endocrinol (Lausanne) 2022; 13:888867. [PMID: 35733766 PMCID: PMC9207331 DOI: 10.3389/fendo.2022.888867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Treatment of heart failure with the angiotensin receptor-neprilysin inhibitor sacubitril/valsartan improved glycemic control in individuals with type 2 diabetes. The relative contribution of neprilysin inhibition versus angiotensin II receptor antagonism to this glycemic benefit remains unknown. Thus, we sought to determine the relative effects of the neprilysin inhibitor sacubitril versus the angiotensin II receptor blocker valsartan on beta-cell function and glucose homeostasis in a mouse model of reduced first-phase insulin secretion, and whether any beneficial effects are additive/synergistic when combined in sacubitril/valsartan. High fat-fed C57BL/6J mice treated with low-dose streptozotocin (or vehicle) were followed for eight weeks on high fat diet alone or supplemented with sacubitril, valsartan or sacubitril/valsartan. Body weight and fed glucose levels were assessed weekly. At the end of the treatment period, insulin release in response to intravenous glucose, insulin sensitivity, and beta-cell mass were determined. Sacubitril and valsartan, but not sacubitril/valsartan, lowered fasting and fed glucose levels and increased insulin release in diabetic mice. None of the drugs altered insulin sensitivity or beta-cell mass, but all reduced body weight gain. Effects of the drugs on insulin release were reproduced in angiotensin II-treated islets from lean C57BL/6J mice, suggesting the insulin response to each of the drugs is due to a direct effect on islets and mechanisms therein. In summary, sacubitril and valsartan each exert beneficial insulinotropic, glycemic and weight-reducing effects in obese and/or diabetic mice when administered alone; however, when combined, mechanisms within the islet contribute to their inability to enhance insulin release.
Collapse
Affiliation(s)
- Nathalie Esser
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
- Laboratory of Immunometabolism and Nutrition, GIGA Infection, Immunity and Inflammation, University of Liège, Liège, Belgium
| | - Christine Schmidt
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Breanne M. Barrow
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Laura Cronic
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Daryl J. Hackney
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Stephen M. Mongovin
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Meghan F. Hogan
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Andrew T. Templin
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Joseph J. Castillo
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Rebecca L. Hull
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Sakeneh Zraika
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
11
|
ZD-2, a novel DPP4 inhibitor, protects islet β-cell and improves glycemic control in high-fat-diet-induced obese mice. Life Sci 2022; 298:120515. [DOI: 10.1016/j.lfs.2022.120515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022]
|
12
|
Efficacy of Sitagliptin on Nonalcoholic Fatty Liver Disease in High-fat-diet-fed Diabetic Mice. Curr Med Sci 2022; 42:513-519. [PMID: 35451807 DOI: 10.1007/s11596-022-2573-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/25/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) is a common cause of clinical liver dysfunction and an important prepathological change of liver cirrhosis. Central obesity, type 2 diabetes mellitus, dyslipidemia, and metabolic syndrome are the major risk factors for NAFLD. Sitagliptin (Sig) is a novel hypoglycemic agent that improves blood glucose levels by increasing the level of active incretin. Sig has been shown to prevent the development of fatty livers in mice on a fructose-rich diet. The purpose of this study was to observe the efficacy of Sig on NAFLD in type 2 diabetic mice. METHODS The diet-induced obesity mouse model was established, and the diabetic mice were screened by an intraperitoneal glucose tolerance trial. The mice were randomly divided into four groups for 8 weeks of intervention: high-fat diet (HFD) group, Sig group, metformin (Met) group, and Sig+Met group. After the intervention, the liver function indexes as well as the blood glucose and blood lipid levels of the mice were measured. In addition, the wet weight of the liver was measured; the pathological sections of the liver tissues were stained to observe the hepatocyte fatty degeneration, inflammation, necrosis, and fibrosis; and the hepatic histological injury was recorded as the NAFLD activity score (NAS). RESULTS Compared with the normal control group, the body weight, liver weight, blood glucose level, insulin resistance (IR), blood lipid level, and transaminase level of the mice in the HFD group were significantly increased, showing typical metabolic syndrome. After treatment with Sig and/or Met, the mice gained less weight, had lower levels of blood glucose, triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and transaminase, and had improved IR compared with the HFD group. The liver pathological NASs in the Sig group (P=0.01), Met group (P=0.028), and Sig+Met group (P<0.001) were lower than those in the HFD group (P<0.05), suggesting that the use of the two drugs alone or in combination can improve the state of liver inflammation. In terms of fibrosis, there was no fibrosis in the control group but there was significant fibrosis in the HFD group (P<0.001). There was no significant difference between the drug intervention groups and the HFD group, indicating that the drug therapy (Sig and/or Met) did not significantly improve the pre-existing fibrosis. CONCLUSION Our experiment proved that Sig can improve NAFLD, including improvement of the serum transaminase level, hepatic pathological inflammation level, and hepatocyte adiposis, suggesting that Sig may play a role by improving glucose and lipid metabolism, reducing the body weight and liver weight, improving insulin sensitivity, and inhibiting fatty liver inflammation. Sig may be a new direction for the treatment of patients with a nonalcoholic fatty liver and diabetes, delaying the progression of NAFLD.
Collapse
|
13
|
Du Y, Li D, Chen J, Li YH, Zhang Z, Hidayat K, Wan Z, Xu JY, Qin LQ. Lactoferrin improves hepatic insulin resistance and pancreatic dysfunctions in high-fat diet and streptozotocin-induced diabetic mice. Nutr Res 2022; 103:47-58. [DOI: 10.1016/j.nutres.2022.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 03/16/2022] [Accepted: 03/20/2022] [Indexed: 11/28/2022]
|
14
|
Esser N, Mongovin SM, Parilla J, Barrow BM, Mundinger TO, Fountaine BS, Larmore MJ, Castillo JJ, Akter R, Hull RL, Zraika S. Neprilysin inhibition improves intravenous but not oral glucose-mediated insulin secretion via GLP-1R signaling in mice with β-cell dysfunction. Am J Physiol Endocrinol Metab 2022; 322:E307-E318. [PMID: 35128957 PMCID: PMC8917916 DOI: 10.1152/ajpendo.00234.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Type 2 diabetes is associated with the upregulation of neprilysin, a peptidase capable of cleaving glucoregulatory peptides such as glucagon-like peptide-1 (GLP-1). In humans, use of the neprilysin inhibitor sacubitril in combination with an angiotensin II receptor blocker was associated with increased plasma GLP-1 levels and improved glycemic control. Whether neprilysin inhibition per se is mediating these effects remains unknown. We sought to determine whether pharmacological neprilysin inhibition on its own confers beneficial effects on glycemic status and β-cell function in a mouse model of reduced insulin secretion, and whether any such effects are dependent on GLP-1 receptor (GLP-1R) signaling. High-fat-fed male wild-type (Glp1r+/+) and GLP-1R knockout (Glp1r-/-) mice were treated with low-dose streptozotocin (STZ) to recapitulate type 2 diabetes-associated β-cell dysfunction, or vehicle as control. Mice were continued on high-fat diet alone or supplemented with the neprilysin inhibitor sacubitril for 8 wk. At the end of the study period, β-cell function was assessed by oral or intravenous glucose-tolerance test. Fasting and fed glucose were significantly lower in wild-type mice treated with sacubitril, although active GLP-1 levels and insulin secretion during oral glucose challenge were unchanged. In contrast, insulin secretion in response to intravenous glucose was significantly enhanced in sacubitril-treated wild-type mice, and this effect was blunted in Glp1r-/- mice. Similarly, sacubitril enhanced insulin secretion in vitro in islets from STZ-treated Glp1r+/+ but not Glp1r-/- mice. Together, our data suggest the insulinotropic effects of pharmacological neprilysin inhibition in a mouse model of β-cell dysfunction are mediated via intra-islet GLP-1R signaling.NEW & NOTEWORTHY The neprilysin inhibitor, sacubitril, improves glycemic status in a mouse model of reduced insulin secretion. Sacubitril enhances intravenous but not oral glucose-mediated insulin secretion. The increased glucose-mediated insulin secretion is GLP-1 receptor-dependent. Neprilysin inhibition does not raise postprandial circulating active GLP-1 levels.
Collapse
Affiliation(s)
- Nathalie Esser
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
| | | | - Jacqueline Parilla
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Breanne M Barrow
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Thomas O Mundinger
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | | | - Megan J Larmore
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Joseph J Castillo
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Rehana Akter
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Rebecca L Hull
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Sakeneh Zraika
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
15
|
Wu M, Carballo-Jane E, Zhou H, Zafian P, Dai G, Liu M, Lao J, Kelly T, Shao D, Gorski J, Pissarnitski D, Kekec A, Chen Y, Previs SF, Scapin G, Gomez-Llorente Y, Hollingsworth SA, Yan L, Feng D, Huo P, Walford G, Erion MD, Kelley DE, Lin S, Mu J. Functionally selective signaling and broad metabolic benefits by novel insulin receptor partial agonists. Nat Commun 2022; 13:942. [PMID: 35177603 PMCID: PMC8854621 DOI: 10.1038/s41467-022-28561-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/17/2022] [Indexed: 01/09/2023] Open
Abstract
Insulin analogs have been developed to treat diabetes with focus primarily on improving the time action profile without affecting ligand-receptor interaction or functional selectivity. As a result, inherent liabilities (e.g. hypoglycemia) of injectable insulin continue to limit the true therapeutic potential of related agents. Insulin dimers were synthesized to investigate whether partial agonism of the insulin receptor (IR) tyrosine kinase is achievable, and to explore the potential for tissue-selective systemic insulin pharmacology. The insulin dimers induced distinct IR conformational changes compared to native monomeric insulin and substrate phosphorylation assays demonstrated partial agonism. Structurally distinct dimers with differences in conjugation sites and linkers were prepared to deliver desirable IR partial agonist (IRPA). Systemic infusions of a B29-B29 dimer in vivo revealed sharp differences compared to native insulin. Suppression of hepatic glucose production and lipolysis were like that attained with regular insulin, albeit with a distinctly shallower dose-response. In contrast, there was highly attenuated stimulation of glucose uptake into muscle. Mechanistic studies indicated that IRPAs exploit tissue differences in receptor density and have additional distinctions pertaining to drug clearance and distribution. The hepato-adipose selective action of IRPAs is a potentially safer approach for treatment of diabetes.
Collapse
MESH Headings
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Alloxan/administration & dosage
- Alloxan/toxicity
- Animals
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- CHO Cells
- Cricetulus
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- HEK293 Cells
- Humans
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/pharmacology
- Insulin/therapeutic use
- Lipolysis/drug effects
- Liver/drug effects
- Liver/metabolism
- Male
- Mice
- Rats
- Receptor, Insulin/agonists
- Recombinant Proteins/pharmacology
- Recombinant Proteins/therapeutic use
- Signal Transduction/drug effects
- Swine
- Swine, Miniature
Collapse
Affiliation(s)
- Margaret Wu
- Merck & Co., Inc., Kenilworth, NJ, 07033, USA
| | | | | | | | - Ge Dai
- Merck & Co., Inc., Kenilworth, NJ, 07033, USA
| | - Mindy Liu
- Merck & Co., Inc., South San Francisco, CA, 94080, USA
| | - Julie Lao
- Merck & Co., Inc., South San Francisco, CA, 94080, USA
| | - Terri Kelly
- Merck & Co., Inc., Kenilworth, NJ, 07033, USA
| | - Dan Shao
- Merck & Co., Inc., South San Francisco, CA, 94080, USA
| | | | | | - Ahmet Kekec
- Merck & Co., Inc., Kenilworth, NJ, 07033, USA
| | - Ying Chen
- Merck & Co., Inc., Kenilworth, NJ, 07033, USA
| | | | | | | | | | - Lin Yan
- Merck & Co., Inc., Kenilworth, NJ, 07033, USA
| | | | - Pei Huo
- Merck & Co., Inc., Kenilworth, NJ, 07033, USA
| | | | | | | | | | - James Mu
- Merck & Co., Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
16
|
Chen L, Yin Z, Qin X, Zhu X, Chen X, Ding G, Sun D, Wu NN, Fei J, Bi Y, Zhang J, Bucala R, Ren J, Zheng Q. CD74 ablation rescues type 2 diabetes mellitus-induced cardiac remodeling and contractile dysfunction through pyroptosis-evoked regulation of ferroptosis. Pharmacol Res 2022; 176:106086. [PMID: 35033649 DOI: 10.1016/j.phrs.2022.106086] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes mellitus (T2D) contributes to sustained inflammation and myopathic changes in the heart although the precise interplay between the two remains largely unknown. This study evaluated the impact of deficiency in CD74, the cognate receptor for the regulatory cytokine macrophage migration inhibitory factor (MIF), in T2D-induced cardiac remodeling and functional responses, and cell death domains involved. WT and CD74-/- mice were fed a high fat diet (60% calorie from fat) for 8 weeks prior to injection of streptozotocin (STZ, 35 mg/kg, i.p., 3 consecutive days) and were maintained for another 8 weeks. KEGG analysis for differentially expressed genes (DEGs) reported gene ontology term related to ferroptosis in T2D mouse hearts. T2D patients displayed elevated plasma MIF levels. Murine T2D exerted overt global metabolic derangements, cardiac remodeling, contractile dysfunction, apoptosis, pyroptosis, ferroptosis and mitochondrial dysfunction, ablation of CD74 attenuated T2D-induced cardiac remodeling, contractile dysfunction, various forms of cell death and mitochondrial defects without affecting global metabolic defects. CD74 ablation rescued T2D-evoked NLRP3-Caspase1 activation and oxidative stress but not dampened autophagy. In vitro evidence depicted that high glucose/high fat (HGHF) compromised cardiomyocyte function and promoted lipid peroxidation, the effects were ablated by inhibitors of NLRP3, pyroptosis, and ferroptosis but not by the mitochondrial targeted antioxidant mitoQ. Recombinant MIF mimicked HGHF-induced lipid peroxidation, GSH depletion and ferroptosis, the effects of which were reversed by inhibitors of MIF, NLRP3 and pyroptosis. Taken together, these data suggest that CD74 ablation protects against T2D-induced cardiac remodeling and contractile dysfunction through NLRP3/pyroptosis-mediated regulation of ferroptosis.
Collapse
MESH Headings
- Adult
- Animals
- Antigens, Differentiation, B-Lymphocyte/genetics
- Cell Line
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Female
- Ferroptosis
- Gene Expression
- Histocompatibility Antigens Class II/genetics
- Humans
- Macrophage Migration-Inhibitory Factors/blood
- Male
- Mice, Knockout
- Middle Aged
- Myocardial Contraction
- Myocardium/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- Oxidative Stress
- Oxygen Consumption
- Pyroptosis
- Rats
- Ventricular Remodeling
- Mice
Collapse
Affiliation(s)
- Lin Chen
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China; The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Zhiqiang Yin
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China; The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Xing Qin
- Department of Cardiology, Xijing Hospital, The Air Force Military Medical University, Xi'an 710032 China
| | - Xiaoying Zhu
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China; The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Xu Chen
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China; The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Gangbing Ding
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China; The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Dong Sun
- Department of Cardiology, Xijing Hospital, The Air Force Military Medical University, Xi'an 710032 China
| | - Ne N Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Juanjuan Fei
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yaguang Bi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Richard Bucala
- Department of Medicine, Yale School of Medicine, New Haven, CT 06520 USA
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle 98195, WA, USA.
| | - Qijun Zheng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China; The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
17
|
McInnes N, Hall S, Hramiak I, Sigal RJ, Goldenberg R, Gupta N, Rabasa-Lhoret R, Braga M, Woo V, Sultan F, Otto R, Smith A, Sherifali D, Liu YY, Gerstein HC, McInnes N, Gerstein HC, Hall S, Blanchard S, Pinder N, Yun Liu Y, McInnes N, Sultan F, Otto R, Smith A, Sherifali D, Gerstein HC, Hramiak I, Paul T, Joy T, Watson M, Driscoll M, Sigal R, Butalia S, Rossiter B, Smith M, Tully V, Goldenberg R, Gupta N, Schlosser R, Sionit D, Talsania T, Huynh J, Birch S, Davdani S, Rabasa-Lhoret R, Bovan D, Raffray M, Braga M, McInnes N, Smith A, Tazzeo T, Otto R, Scott K, Hiltz Mackenzie K, Woo V, Berard L, Mandock C, Anderlic T. Remission of Type 2 Diabetes Following a Short-term Intensive Intervention With Insulin Glargine, Sitagliptin, and Metformin: Results of an Open-label Randomized Parallel-Design Trial. Diabetes Care 2022; 45:178-185. [PMID: 34728531 DOI: 10.2337/dc21-0278] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 09/29/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The aim of the study was to evaluate remission of type 2 diabetes following a short-term intervention with insulin glargine, sitagliptin/metformin, and lifestyle approaches. RESEARCH DESIGN AND METHODS In this open multicenter trial, 102 patients with type 2 diabetes were randomized to 1) a 12-week intervention with sitagliptin/metformin, insulin glargine, and lifestyle therapy or 2) control group. Participants with HbA1c <7.3% (<56 mmol/mol) at 12 weeks were asked to stop diabetes medications and were followed for evidence of relapse over 52 weeks. Diabetes relapse criteria included HbA1c ≥6.5% (≥48 mmol/mol), ≥50% of capillary glucose readings >10 mmol/L over 1 week, and reinitiation of diabetes medications with or without abnormal fasting plasma glucose (FPG) or 2-h plasma glucose on an oral glucose tolerance test (OGTT). Time-to-relapse analysis was conducted to compare the treatment groups with (primary analysis) and without (supplementary analysis) FPG/OGTT relapse criteria. RESULTS With the FPG/OGTT relapse criteria included, the hazard ratio (HR) of relapse was 0.72 (95% CI 0.47-1.10) in the intervention group compared with the control group (primary analysis), and the number of participants remaining in remission was not significantly different between treatment groups at 24, 36, 48, and 64 weeks. In the supplementary analyses without these criteria, HR of relapse was 0.60 (95% CI 0.39-0.95), and the number of participants remaining in remission was significantly higher (26 vs. 10%) in the intervention group at 36 weeks. CONCLUSIONS Although our primary outcome was not statistically significant, the tested approach deserves further study with further optimization of its components.
Collapse
Affiliation(s)
- Natalia McInnes
- 1Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,2Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada.,3Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Stephanie Hall
- 2Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Irene Hramiak
- 4Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Ronald J Sigal
- 5Departments of Medicine, Cardiac Sciences and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Nikhil Gupta
- 6LMC Diabetes and Endocrinology, Concord, Ontario, Canada
| | - Remi Rabasa-Lhoret
- 7Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Manoela Braga
- 1Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Vincent Woo
- 8Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Farah Sultan
- 3Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Rose Otto
- 1Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ada Smith
- 1Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Diana Sherifali
- 9School of Nursing, McMaster University, Hamilton, Ontario, Canada
| | - Yan Yun Liu
- 2Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Hertzel C Gerstein
- 1Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,2Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada.,3Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Fu Y, Zhang F, Liu Z, Zhao Q, Xue Y, Shen Q. Improvement of diabetes-induced metabolic syndrome by millet prolamin is associated with changes in serum metabolomics. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
Torrecillas-Baena B, Gálvez-Moreno MÁ, Quesada-Gómez JM, Dorado G, Casado-Díaz A. Influence of Dipeptidyl Peptidase-4 (DPP4) on Mesenchymal Stem-Cell (MSC) Biology: Implications for Regenerative Medicine - Review. Stem Cell Rev Rep 2021; 18:56-76. [PMID: 34677817 DOI: 10.1007/s12015-021-10285-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Dipeptidyl peptidase IV (DPP4) is a ubiquitous protease that can be found in membrane-anchored or soluble form. Incretins are one of the main DPP4 substrates. These hormones regulate glucose levels, by stimulating insulin secretion and decreasing glucagon production. Because DPP4 levels are high in diabetes, DPP4 inhibitor (DPP4i) drugs derived from gliptin are widespread used as hypoglycemic agents for its treatment. However, as DPP4 recognizes other substrates such as chemokines, growth factors and neuropeptides, pleiotropic effects have been observed in patients treated with DPP4i. Several of these substrates are part of the stem-cell niche. Thus, they may affect different physiological aspects of mesenchymal stem-cells (MSC). They include viability, differentiation, mobilization and immune response. MSC are involved in tissue homeostasis and regeneration under both physiological and pathological conditions. Therefore, such cells and their secretomes have a high clinical potential in regenerative medicine. In this context, DPP4 activity may modulate different aspects of MSC regenerative capacity. Therefore, the aim of this review is to analyze the effect of different DPP4 substrates on MSC. Likewise, how the regulation of DPP4 activity by DPP4i can be applied in regenerative medicine. That includes treatment of cardiovascular and bone pathologies, cutaneous ulcers, organ transplantation and pancreatic beta-cell regeneration, among others. Thus, DPP4i has an important clinical potential as a complement to therapeutic strategies in regenerative medicine. They involve enhancing the differentiation, immunomodulation and mobilization capacity of MSC for regenerative purposes.
Collapse
Affiliation(s)
- Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Gabriel Dorado
- Dep. Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, CIBERFES, 14071, Córdoba, Spain
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain.
| |
Collapse
|
20
|
Doolittle ML, Monroe DG, Farr JN, Khosla S. The role of senolytics in osteoporosis and other skeletal pathologies. Mech Ageing Dev 2021; 199:111565. [PMID: 34499959 DOI: 10.1016/j.mad.2021.111565] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/20/2021] [Accepted: 09/03/2021] [Indexed: 11/26/2022]
Abstract
The skeletal system undergoes irreversible structural deterioration with aging, leading to increased fracture risk and detrimental changes in mobility, posture, and gait. This state of low bone mass and microarchitectural changes, diagnosed as osteoporosis, affects millions of individuals worldwide and has high clinical and economic burdens. Recently, pre-clinical studies have linked the onset of age-related bone loss with an accumulation of senescent cells in the bone microenvironment. These senescent cells appear to be causal to age-related bone loss, as targeted clearance of these cells leads to improved bone mass and microarchitecture in old mice. Additionally, other pathologies leading to bone loss that result from DNA damage, such as cancer treatments, have shown improvements after clearance of senescent cells. The development of new therapies that clear senescent cells, termed "senolytics", is currently underway and may allow for the modulation of bone loss that results from states of high senescent cell burden, such as aging.
Collapse
Affiliation(s)
- Madison L Doolittle
- Kogod Center on Aging and Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, 55905, United States
| | - David G Monroe
- Kogod Center on Aging and Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, 55905, United States
| | - Joshua N Farr
- Kogod Center on Aging and Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, 55905, United States
| | - Sundeep Khosla
- Kogod Center on Aging and Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, 55905, United States.
| |
Collapse
|
21
|
Fushimi Y, Obata A, Sanada J, Nogami Y, Ikeda T, Yamasaki Y, Obata Y, Shimoda M, Nakanishi S, Mune T, Kaku K, Kaneto H. Early combination therapy of empagliflozin and linagliptin exerts beneficial effects on pancreatic β cells in diabetic db/db mice. Sci Rep 2021; 11:16120. [PMID: 34373487 PMCID: PMC8352868 DOI: 10.1038/s41598-021-94896-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Effects of combination therapy of dipeptidyl peptidase-4 (DPP-4) inhibitor and sodium-glucose co-transporter 2 (SGLT2) inhibitor on β-cells are still unclear, although combination agent of these two drugs has become common in clinical practice. Therefore, we aimed to elucidate the effects of DPP-4 inhibitor and/or SGLT2 inhibitor on β-cell mass and function and compared their effects between in an early and advanced phase of diabetes. We used 7-week-old db/db mice as an early phase and 16-week-old mice as an advanced phase and treated them for 2 weeks with oral administration of linagliptin, empagliflozin, linagliptin + empagliflozin (L + E group), and 0.5% carboxymethylcellulose (Cont group). Blood glucose levels in Empa and L + E group were significantly lower than Cont group after treatment. In addition, β-cell mass in L + E group was significantly larger than Cont group only in an early phase, accompanied by increased Ki67-positive β-cell ratio. In isolated islets, mRNA expression levels of insulin and its transcription factors were all significantly higher only in L + E group in an early phase. Furthermore, mRNA expression levels related to β-cell differentiation and proliferation were significantly increased only in L + E group in an early phase. In conclusion, combination of DPP-4 inhibitor and SGLT2 inhibitor exerts more beneficial effects on β-cell mass and function, especially in an early phase of diabetes rather than an advanced phase.
Collapse
Affiliation(s)
- Yoshiro Fushimi
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Atsushi Obata
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan.
| | - Junpei Sanada
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Yuka Nogami
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Tomoko Ikeda
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Yuki Yamasaki
- Department of Clinical Nutrition, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, 701-0193, Japan
| | - Yoshiyuki Obata
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Masashi Shimoda
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Shuhei Nakanishi
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Tomoatsu Mune
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Kohei Kaku
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Hideaki Kaneto
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan.
| |
Collapse
|
22
|
Pinheiro MM, Pinheiro FMM, Diniz SN, Fabbri A, Infante M. Combination of vitamin D and dipeptidyl peptidase-4 inhibitors (VIDPP-4i) as an immunomodulation therapy for autoimmune diabetes. Int Immunopharmacol 2021; 95:107518. [PMID: 33756226 DOI: 10.1016/j.intimp.2021.107518] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) and latent autoimmune diabetes in adults (LADA) represent the most common types of autoimmune diabetes and are characterized by different age of onset, degrees of immune-mediated destruction of pancreatic beta cells and rates of disease progression towards insulin dependence. Several immunotherapies aimed to counteract autoimmune responses against beta cells and preserve beta-cell function are currently being investigated, particularly in T1D. Preliminary findings suggest a potential role of combination therapy with vitamin D and dipeptidyl peptidase-4 (DPP-4) inhibitors (VIDPP-4i) in preserving beta-cell function in autoimmune diabetes. This manuscript aims to provide a comprehensive overview of the immunomodulatory properties of vitamin D and DPP-4 inhibitors, as well as the rationale for investigation of their combined use as an immunomodulation therapy for autoimmune diabetes.
Collapse
Affiliation(s)
- Marcelo Maia Pinheiro
- UNIVAG, University Center, Dom Orlando Chaves Ave, 2655 - Cristo Rei, Várzea Grande, 78118-000 Mato Grosso, Brazil; Universidade Anhanguera de São Paulo - SP, 3305, Raimundo Pereira de Magalhães Ave., Pirituba, São Paulo, 05145-200 São Paulo, Brazil.
| | - Felipe Moura Maia Pinheiro
- Hospital de Base, Faculdade de Medicina de São José do Rio Preto FAMERP - SP, 5546, Brigadeiro Faria Lima Ave, Vila São Pedro, São José do Rio Preto, 15015-500 São Paulo, Brazil
| | - Susana Nogueira Diniz
- Universidade Anhanguera de São Paulo - SP, 3305, Raimundo Pereira de Magalhães Ave., Pirituba, São Paulo, 05145-200 São Paulo, Brazil
| | - Andrea Fabbri
- Diabetes Research Institute Federation (DRIF), Division of Endocrinology and Diabetes, CTO Andrea Alesini Hospital, ASL Roma 2, Department of Systems Medicine, University of Rome Tor Vergata, Via San Nemesio 21, 00145 Rome, Italy
| | - Marco Infante
- Diabetes Research Institute Federation (DRIF), Division of Endocrinology and Diabetes, CTO Andrea Alesini Hospital, ASL Roma 2, Department of Systems Medicine, University of Rome Tor Vergata, Via San Nemesio 21, 00145 Rome, Italy; UniCamillus, Saint Camillus International University of Health Sciences, Via di Sant'Alessandro, 8, 00131 Rome, Italy; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Via San Nemesio 21, 00145 Rome, Italy.
| |
Collapse
|
23
|
Chao J, Cheng HY, Chang ML, Huang SS, Liao JW, Cheng YC, Peng WH, Pao LH. Gallic Acid Ameliorated Impaired Lipid Homeostasis in a Mouse Model of High-Fat Diet-and Streptozotocin-Induced NAFLD and Diabetes through Improvement of β-oxidation and Ketogenesis. Front Pharmacol 2021; 11:606759. [PMID: 33643038 PMCID: PMC7907449 DOI: 10.3389/fphar.2020.606759] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/24/2020] [Indexed: 12/23/2022] Open
Abstract
Gallic acid (GA) is a simple polyphenol found in food and traditional Chinese medicine. Here, we determined the effects of GA administration in a combined mouse model of high-fat diet (HFD)-induced obesity and low-dose streptozotocin (STZ)-induced hyperglycemia, which mimics the concurrent non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes pathological condition. By combining the results of physiological assessments, pathological examinations, metabolomic studies of blood, urine, liver, and muscle, and measurements of gene expression, we attempted to elucidate the efficacy of GA and the underlying mechanism of action of GA in hyperglycemic and dyslipidemic mice. HFD and STZ induced severe diabetes, NAFLD, and other metabolic disorders in mice. However, the results of liver histopathology and serum biochemical examinations indicated that daily GA treatment alleviated the high blood glucose levels in the mice and decelerated the progression of NAFLD. In addition, our results show that the hepatoprotective effect of GA in diabetic mice occurs in part through a partially preventing disordered metabolic pathway related to glucose, lipids, amino acids, purines, and pyrimidines. Specifically, the mechanism responsible for alleviation of lipid accumulation is related to the upregulation of β-oxidation and ketogenesis. These findings indicate that GA alleviates metabolic diseases through novel mechanisms.
Collapse
Affiliation(s)
- Jung Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Hao-Yuan Cheng
- Department of Nursing, Chung-Jen Junior College of Nursing, Health Sciences and Management, Chia-Yi, Taiwan
| | - Ming-Ling Chang
- Division of Hepatology, Department of Gastroenterology and Hepatology, Liver Research Center, Chang Gung Memorial Hospital, Linko, Taiwan
| | | | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, Taiwan
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| | - Wen-Huang Peng
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Li-Heng Pao
- Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, and Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linko, Taiwan
| |
Collapse
|
24
|
Abdalla MA, Deshmukh H, Atkin S, Sathyapalan T. The potential role of incretin-based therapies for polycystic ovary syndrome: a narrative review of the current evidence. Ther Adv Endocrinol Metab 2021; 12:2042018821989238. [PMID: 33552465 PMCID: PMC7844452 DOI: 10.1177/2042018821989238] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/03/2021] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age. Metabolic consequences associated with PCOS include, but are not limited to, insulin resistance (IR), type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). This narrative review aims to provide a comprehensive overview of the potential therapeutic roles of the incretin-based therapies in the management of PCOS. METHODS We performed a systematic search of databases including PubMed, MEDLINE and EMBASE up to 1 October 2020. We developed a search string of medical subject headings (MeSH) including the terms PCOS, incretin mimetics, glucagon-like peptide-1 (GLP-1), glucagon-like peptide-1 receptor antagonists (GLP-1 RAs), liraglutide, exenatide, semaglutide, dipeptidyl peptidase-4 (DPP-4) inhibitors, combined with IR, testosterone and sex hormone-binding globulin (SHBG). RESULTS We identified 854 relevant articles and, after the initial screening, eight interventional animal studies, one observational animal study, 14 interventional human studies, two case-control studies and one systematic review were included. These studies showed the potential significant roles of GLP-1 RAs and DPP-4 inhibitors in the management of PCOS, with significant improvements in the metabolic parameters, including substantial weight reduction and improved insulin sensitivity. These agents also improved the hormonal parameters through decreased free androgen and increased SHBG. Moreover, they improved menstrual regularity, increased fertility with enhanced ovulation and pregnancy in obese women with PCOS. CONCLUSION GLP-1 RAs and DPP-4 inhibitors have a promising therapeutic role in PCOS; however, larger clinical trials are needed to establish the role of incretin-based therapies in the management of PCOS.
Collapse
Affiliation(s)
- Mohammed Altigani Abdalla
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Harshal Deshmukh
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Stephen Atkin
- School of Postgraduate Studies and Research, RCSI Medical University of Bahrain, Kingdom of Bahrain
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| |
Collapse
|
25
|
Bellis A, Mauro C, Barbato E, Ceriello A, Cittadini A, Morisco C. Stress-Induced Hyperglycaemia in Non-Diabetic Patients with Acute Coronary Syndrome: From Molecular Mechanisms to New Therapeutic Perspectives. Int J Mol Sci 2021; 22:E775. [PMID: 33466656 PMCID: PMC7828822 DOI: 10.3390/ijms22020775] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 01/08/2023] Open
Abstract
Stress-induced hyperglycaemia (SIH) at hospital admission for acute coronary syndrome is associated with poor outcome, especially in patients without known diabetes. Nevertheless, insulin treatment in these subjects was not correlated with the reduction of mortality. This is likely due to the fact that SIH in the context of an acute coronary syndrome, compared to that in known diabetes, represents an epiphenomenon of other pathological conditions, such as adrenergic and renin-angiotensin system over-activity, hyperglucagonaemia, increase of circulating free fatty acids and pancreatic beta-cell dysfunction, which are not completely reversed by insulin therapy and so worsen the prognosis. Thus, SIH may be considered not only as a biomarker of organ damage, but also as an indicator of a more complex therapeutic strategy in these subjects. The aim of this review is to analyse the molecular mechanisms by which SIH may favour a worse prognosis in non-diabetic patients with acute coronary syndrome and identify new therapeutic strategies, in addition to insulin therapy, for a more appropriate treatment and improved outcomes.
Collapse
Affiliation(s)
- Alessandro Bellis
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica-Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy; (A.B.); (C.M.)
| | - Ciro Mauro
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica-Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy; (A.B.); (C.M.)
| | - Emanuele Barbato
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, 80131 Napoli, Italy;
| | - Antonio Ceriello
- Department of Cardiovascular and Metabolic Diseases, IRCCS Multimedica, Sesto San Giovanni, 20099 Milan, Italy;
| | - Antonio Cittadini
- Dipartimento di Scienze Mediche Traslazionali, Università Federico II, 80131 Napoli, Italy;
| | - Carmine Morisco
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, 80131 Napoli, Italy;
| |
Collapse
|
26
|
Lee S, Lee S, Jang IJ, Yu KS, Rhee SJ. Electronic medical records-based comparison of glycemic control efficacy between sulfonylureas and dipeptidyl peptidase-4 inhibitors added on to metformin monotherapy in patients with type 2 diabetes. Transl Clin Pharmacol 2021; 28:199-207. [PMID: 33425803 PMCID: PMC7781806 DOI: 10.12793/tcp.2020.28.e21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 11/19/2022] Open
Abstract
Sulfonylurea (SU) and dipeptidyl peptidase-4 (DPP-4) inhibitors are most common secondary agents that are added to metformin monotherapy. Real-world studies have become increasingly important in providing evidence of treatment effectiveness in clinical practice and real-world data could help appropriate therapeutic information. Therefore, this study aims to compare the glycemic effectiveness of SU and DPP-4 inhibitors, which are added to metformin monotherapy in real clinical practice using electronic medical record (EMR) data. EMR data of type 2 diabetes patients treated at Seoul National University Hospital from December 2002 to December 2012 were retrieved and analyzed. The patients were divided into three groups: patients who maintained metformin monotherapy (M), and patients who added SU (MS) or DPP-4 inhibitors (MD) to metformin monotherapy. The mean change in HbA1c level, the proportion of patients achieving the HbA1c target < 7.0%, proportion of patients with treatment failure, and probability of treatment failure occurrence and changes in prescription were evaluated to compare glycemic control efficacy between SU and DPP-4 inhibitors. The MS showed significantly greater reduction in the Hb1Ac level than MD. The proportion of patients achieving HbA1c < 7.0% is higher in MD, whereas the proportion of patients with treatment failure was greater in MS. The probability of the treatment failure and probability of changes in the prescription were lower in MD than MS with hazard ratio of 0.499 and 0.579, respectively. In conclusion, this real-world study suggested that DPP-4 inhibitors are expected to show more durable glycemic control efficacy than SU in long-term use.
Collapse
Affiliation(s)
- Suhrin Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - Su-Jin Rhee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| |
Collapse
|
27
|
Li D, Zou H, Yin P, Li W, He J, Wang S, Huang L, Shao S, Chen Y, Yang Y, Yu X. Durability of glycaemic control in type 2 diabetes: A systematic review and meta-analysis for its association with body weight changes. Diabetes Obes Metab 2021; 23:208-217. [PMID: 33016522 DOI: 10.1111/dom.14217] [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: 06/09/2020] [Revised: 08/26/2020] [Accepted: 09/20/2020] [Indexed: 10/23/2022]
Abstract
AIMS To analyse quantitatively the association between the durability of glycaemic control and body weight changes during treatment. MATERIALS AND METHODS This study adhered to an appropriate methodology according to Meta-analysis of Observational Studies in Epidemiology (MOOSE) guidelines. Studies with follow-ups >12 months, and final and intermediate assessments of haemoglobin A1c (HbA1c) and body weight were included. Four outcomes assessing therapeutic durability were extracted and synthesized using Stata statistical software, including changes in HbA1c, goal-achievement rate, failure rate and coefficient of failure (CoF). RESULTS After 8.9 months of treatment, HbA1c levels declined from 8.03% [95% confidence interval (CI), 7.91-8.15; I2 = 99.2%] to 7.15% (95% CI, 7.02-7.27; I2 = 99.4%) and then gradually increased up to 7.72% (95% CI, 7.50-7.94; I2 = 99.0%) 5 years later. The goal-achievement rate decreased from 54.8% (after 1 year of treatment) to 19.4% 5 years later. The CoF was 0.123 ± 0.022%/year (P < .001). After stratification, the CoFs were 0.224 ± 0.025%/year (P < .001) for weight gain, 0.137 ± 0.034%/year (P < .001) for neutral weight and -0.024 ± 0.032%/year (P = .450) for weight loss. After stratification by treatment approaches, the CoFs were 0.45%/year for insulin, 0.43%/year for sulphonylurea, 0.34%/year for thiazolidinediones, 0.29%/year for metformin, 0.16% for glucagon-like polypeptide-1 receptor agonists, 0.12% for surgery, -0.03% for sodium-glucose cotransporter-2 inhibitors and -0.21% for dipeptidyl peptidase-IV inhibitors. CONCLUSION Modest weight loss with a goal of 2-3% of body weight should be recommended to improve therapeutic durability and prevent beta-cell deterioration.
Collapse
Affiliation(s)
- Danpei Li
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - HuaJie Zou
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjun Li
- Computer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyu He
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuyun Wang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Huang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiying Shao
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Yang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuefeng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Li P, Fan C, Cai Y, Fang S, Zeng Y, Zhang Y, Lin X, Zhang H, Xue Y, Guan M. Transplantation of brown adipose tissue up-regulates miR-99a to ameliorate liver metabolic disorders in diabetic mice by targeting NOX4. Adipocyte 2020; 9:57-67. [PMID: 32000567 PMCID: PMC6999837 DOI: 10.1080/21623945.2020.1721970] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), main cause of liver damage, is inextricably linked to diabetes. However, there is no specific means to improve the pathology of fatty liver in diabetic patients. Brown adipose tissue (BAT) is an important endocrine organ that secretes adipokines and microRNAs (miRNAs) involved in systemic metabolic regulation. To investigate the effects of BAT transplantation on liver lipid metabolism in diabetic mice, we transplanted BAT from male donor mice into diabetic mice induced by streptozotocin (STZ) combined with high-fat diet (HFD). At 10 weeks after transplantation, BAT transplantation significantly decreased the blood glucose and lipid, downregulated FAS, CD36, Scd1, ACCα, NOX2, NOX4, TGF-β1, FN and COL-1, up-regulated Nrf2, reversed the pathological changes of liver and increased the circulating miR-99a in diabetic mice. To verify whether circulating miR-99a improves oxidative stress by targeting inhibition of NOX4, we used 0.4mM palmitic acid (PA) to treat the LO2 cells. The expression of NOX4 protein was significantly decreased after transfection with miR-99a mimic, and increased after transfection with miR-99a inhibitor. Luciferase reporter assay confirmed that miR-99a could target NOX4 mRNA. These findings clarify the role of miR-99a and NOX4 in liver beneficial effect of BAT transplantation in diabetic mice.
Collapse
Affiliation(s)
- Ping Li
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Endocrinology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Cunxia Fan
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Endocrinology and Metabolism, Hainan General Hospital, Haikou, Hainan, China
| | - Yingying Cai
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shu Fang
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanmei Zeng
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yudan Zhang
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaochun Lin
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongbin Zhang
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yaoming Xue
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Meiping Guan
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Fu Y, Yin R, Liu Z, Niu Y, Guo E, Cheng R, Diao X, Xue Y, Shen Q. Hypoglycemic Effect of Prolamin from Cooked Foxtail Millet ( Setaria italic) on Streptozotocin-Induced Diabetic Mice. Nutrients 2020; 12:E3452. [PMID: 33187155 PMCID: PMC7696583 DOI: 10.3390/nu12113452] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Millet proteins have been demonstrated to possess glucose-lowering and lipid metabolic disorder modulation functions against diabetes; however, the molecular mechanisms underlying their anti-diabetic effects remain unclear. The present study aimed to investigate the hypoglycemic effect of prolamin from cooked foxtail millet (PCFM) on type 2 diabetic mice, and explore the gut microbiota and serum metabolic profile changes that are associated with diabetes attenuation by PCFM. Our diabetes model was established using a high-fat diet combined with streptozotocin before PCFM or saline was daily administrated by gavage for 5 weeks. The results showed that PCFM ameliorated glucose metabolism disorders associated with type 2 diabetes. Furthermore, the effects of PCFM administration on gut microbiota and serum metabolome were investigated. 16S rRNA gene sequencing analysis indicated that PCFM alleviated diabetes-related gut microbiota dysbiosis in mice. Additionally, the serum metabolomics analysis revealed that the metabolite levels disturbed by diabetes were partly altered by PCFM. Notably, the decreased D-Glucose level caused by PCFM suggested that its anti-diabetic potential can be associated with the activation of glycolysis and the inhibition of gluconeogenesis, starch and sucrose metabolism and galactose metabolism. In addition, the increased serotonin level caused by PCFM may stimulate insulin secretion by pancreatic β-cells, which contributed to its hypoglycemic effect. Taken together, our research demonstrated that the modulation of gut microbiota composition and the serum metabolomics profile was associated with the anti-diabetic effect of PCFM.
Collapse
Affiliation(s)
- Yongxia Fu
- Key Laboratory of Plant Protein and Grain Processing, National Engineering Research Center for Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.F.); (R.Y.); (Z.L.); (Y.X.)
| | - Ruiyang Yin
- Key Laboratory of Plant Protein and Grain Processing, National Engineering Research Center for Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.F.); (R.Y.); (Z.L.); (Y.X.)
| | - Zhenyu Liu
- Key Laboratory of Plant Protein and Grain Processing, National Engineering Research Center for Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.F.); (R.Y.); (Z.L.); (Y.X.)
| | - Yan Niu
- Shan Xi Dongfang Wuhua Agricultural Technology Co. Ltd., Datong 037000, China;
| | - Erhu Guo
- Research Institute of Millet, Shanxi Academy of Agricultural Sciences, Taiyuan 030031, China;
| | - Ruhong Cheng
- Research Institute of Millet, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang 050035, China;
| | - Xianmin Diao
- Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China;
| | - Yong Xue
- Key Laboratory of Plant Protein and Grain Processing, National Engineering Research Center for Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.F.); (R.Y.); (Z.L.); (Y.X.)
| | - Qun Shen
- Key Laboratory of Plant Protein and Grain Processing, National Engineering Research Center for Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.F.); (R.Y.); (Z.L.); (Y.X.)
| |
Collapse
|
30
|
Li J, Xie F, Zhu X, Hu H, Fang X, Huang Z, Liu Z, Wu S. Polysaccharides from the Fleshy Fruits of Camellia oleifera Attenuate the Kidney Injury in High-Fat Diet/Streptozotocin-Induced Diabetic Mice. J Med Food 2020; 23:1275-1286. [PMID: 33090944 DOI: 10.1089/jmf.2020.4717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The fleshy fruits of Camellia oleifera Abel are the immature fruits of C. oleifera, which are infected by Exobasidium vexans Massee and then turn to be intumescent and hollowed. They contain rich trace elements and vitamin C and are eaten directly as wild fruits in the Chinese countryside. Recent studies report that C. oleifera has anti-inflammatory and antioxidative effects. The current study, for the first time, evaluates the renal protective capacity of polysaccharides from the fleshy fruits of C. oleifera (CFFP) in streptozotocin-induced diabetic mice fed high-fat diets. The diabetic mice were orally administered CFFP for 3 months to evaluate the renoprotective function of CFFP. Our results indicated that 250 mg/kg CFFP significantly alleviated diabetes-induced renal injury by decreasing serum creatine, blood urea nitrogen levels, the kidney/body weight ratio, expression of fibronectin and collagen, as well as the secretion of tumor necrosis factor-α and interleukin-6. Additionally, 250 mg/kg CFFP could significantly ameliorate renal oxidative stress through increasing glutathione levels and lowering malondialdehyde contents. We confirmed that CFFP could exert antioxidative, anti-inflammatory, and antifibrosis activities. CFFP might be a potential therapeutic agent, and the fleshy fruits of C. oleifera might be a diet therapy for diabetic patients in the future.
Collapse
Affiliation(s)
- Jialin Li
- School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Fuhua Xie
- School of Basic Medicine, and Gannan Medical University, Ganzhou, China
| | - Xiuzhi Zhu
- School of Basic Medicine, and Gannan Medical University, Ganzhou, China
| | - Haibo Hu
- School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Xiansong Fang
- The First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Zhiwei Huang
- School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Zhiping Liu
- School of Basic Medicine, and Gannan Medical University, Ganzhou, China
| | - Suzhen Wu
- School of Basic Medicine, and Gannan Medical University, Ganzhou, China
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Senescent cells are now known to accumulate in multiple tissues with aging and through their inflammation (the senescence-associated secretory phenotype, SASP) contribute to aging and chronic diseases. Here, we review the roles of senescent osteocytes in the context of bone loss. RECENT FINDINGS Numerous studies have established that senescent osteocytes accumulate in the bone microenvironment with aging in mice and in humans. Moreover, at least in mice, elimination of senescent cells results in attenuation of age-related bone loss. Osteocyte senescence also occurs in response to other cellular stressors, including radiotherapy, chemotherapy, and metabolic dysfunction, where it appears to mediate skeletal deterioration. Osteocyte senescence is linked to bone loss associated with aging and other conditions. Senescent osteocytes are potential therapeutic targets to alleviate skeletal dysfunction. Additional studies better defining the underlying mechanisms as well as translating these exciting findings from mouse models to humans are needed.
Collapse
Affiliation(s)
- Joshua N Farr
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Division of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Japneet Kaur
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Madison L Doolittle
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sundeep Khosla
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
32
|
Juang JH, Chen CY, Kao CW, Huang YW, Chiu TY, Chen CT. Implanted islet mass influences the effects of dipeptidyl peptidase-IV inhibitor LAF237 on transplantation outcomes in diabetic mice. Biomed J 2020; 44:S210-S217. [PMID: 35300943 PMCID: PMC9068567 DOI: 10.1016/j.bj.2020.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/10/2020] [Accepted: 10/06/2020] [Indexed: 01/06/2023] Open
Abstract
Background Previous studies showed inconsistent Results of the effects of dipeptidyl peptidase (DPP)-IV inhibitors on syngeneic mouse islet transplantation. We hypothesized that the implanted islet numbers are critical for the effects of DPP-IV inhibitors on the outcomes of transplantation. Methods One hundred and fifty or three hundred islets were syngeneically transplanted under the renal capsule of each streptozocin-diabetic C57BL/6 mouse and recipients were then treated without or with LAF237 (10 mg/kg/day, po) for 6 weeks. After transplantation, recipients’ blood glucose, body weight and intraperitoneal glucose tolerance test (IPGTT) were followed-up periodically. The graft was removed for the measurement of β-cell mass at 6 weeks. Results In recipients with 150 islets, it was not significantly different between the LAF237- treated group (n = 14) and control group (n = 14) in terms of the blood glucose, body weight, glucose tolerance at 2, 4 and 6 weeks or the graft β-cell mass at 6 weeks. In contrast, in recipients with 300 islets, the LAF237-treated group (n = 24) did have a lower area under the curve of the IPGTT at 4 weeks (p = 0.0237) and 6 weeks (p = 0.0113) as well as more graft β-cell mass at 6 weeks (0.655 ± 0.008 mg vs. 0.435 ± 0.006 mg, p = 0.0463) than controls (n = 24). Conclusions Our findings revealed 6-week treatment of LAF237 improves glucose tolerance and increases graft β-cell mass in diabetic mice transplanted with a sufficient number but not a marginal number of islets. These indicate that the effects of DPP-IV inhibitors are influenced by the implanted islet mass.
Collapse
Affiliation(s)
- Jyuhn-Huarng Juang
- Division of Endocrinology and Metabolism, Center for Tissue Engineering, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Chen-Yi Chen
- Division of Endocrinology and Metabolism, Center for Tissue Engineering, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chen-Wei Kao
- Division of Endocrinology and Metabolism, Center for Tissue Engineering, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yu-Wen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
33
|
Comparisons between dipeptidyl peptidase-4 inhibitors and other classes of hypoglycemic drugs using two distinct biomarkers of pancreatic beta-cell function: A meta-analysis. PLoS One 2020; 15:e0236603. [PMID: 32706828 PMCID: PMC7380634 DOI: 10.1371/journal.pone.0236603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/08/2020] [Indexed: 01/09/2023] Open
Abstract
Background and objective Dipeptidyl peptidase-4 (DPP-4) inhibitors have been suggested to have pancreatic beta-cell preserving effect according to studies using homeostatic model of assessment for beta-cell function (HOMA-β). However, whether HOMA-β is a suitable biomarker for comparisons between hypoglycemic drugs with different mechanisms of action remains unclear. Therefore, we conducted a meta-analysis to compare the effects of DPP-4 inhibitors and other classes of hypoglycemic drugs on HOMA-β and proinsulin-to-insulin ratio (PIR). Methods We searched MEDLINE, CENTRAL, and Ichushi-web for the period of 1966 to May 2020. We collected randomized, controlled clinical trials in patients with type 2 diabetes mellitus comparing DPP-4 inhibitors and other classes of hypoglycemic agents [α-glucosidase inhibitors (α-GIs), glucagon-like peptide-1 (GLP-1) analogues, metformin, sodium-glucose cotransporter 2 (SGLT2) inhibitors, sulfonylureas, or thiazolidinediones]. Weighted mean differences and 95% confidence intervals of changes in HOMA-β or PIR during study periods were calculated for pairwise comparisons. Results Thirty-seven and 21 relevant trials were retrieved for comparisons of HOMA-β and PIR, respectively. HOMA-β and PIR consistently showed superiority of DPP-4 inhibitors compared with α-GIs. Both biomarkers consistently supported inferiority of DPP-4 inhibitors compared with GLP-1 analogues. However, PIR showed inferiority of DPP-4 inhibitors compared with metformin, and superiority compared with SGLT2 inhibitors, whereas HOMA-β showed no significant differences between DPP-4 inhibitors and the two other agents. Conclusion DPP-4 inhibitors appear to be superior to α-GIs but inferior to GLP-1 analogues in preservation of beta-cell function assessed by either HOMA-β or PIR. DPP-4 inhibitors seem to be superior to SGLT2 inhibitors but inferior to metformin on islet function assessed only by PIR. Because HOMA-β and PIR may indicate different aspects of beta-cell function, results of beta-cell function preserving effects of hypoglycemic agents should be interpreted with caution.
Collapse
|
34
|
Le TD, Nguyen NTP, Nguyen ST, Tran HTT, Nguyen LTH, Duong HH, Nguyen HM, Do BN. Sitagliptin Increases Beta-Cell Function and Decreases Insulin Resistance in Newly Diagnosed Vietnamese Patients with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2020; 13:2119-2127. [PMID: 32606870 PMCID: PMC7310979 DOI: 10.2147/dmso.s255071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/29/2020] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION To investigate effects of Sitagliptin on the enhancement of beta-cell function, reducing insulin resistance, serum glucagon like peptide-1 (GLP-1) concentrations and blood glucose in patients with type 2 diabetes mellitus (T2D) and suggest one of the underlying mechanisms on beta-cell function and insulin resistance. PATIENTS AND METHODS This was a cross-sectional and observational study in comparison to the control group. A study population of 44 newly diagnosed patients with T2D treated with Sitagliptin with a dose of 100 mg/day for 3 months was analyzed to compare 52 healthy participants. Indices for beta-cell function, peripheral insulin sensitivity, and insulin resistance were calculated with homeostasis model assessment 2 (HOMA2) calculator and compared. Serum GLP-1 concentrations were analyzed, and regression analysis was conducted to find the correlations between GLP-1 and beta-cell function and insulin resistance. RESULTS Newly diagnosed patients with T2D witnessed a significant reduction in beta-cell function, serum GLP-1 concentrations at the time of diagnosis. After treatment with Sitagliptin 100 mg/day, they achieved significant improvements in beta-cell function, peripheral insulin sensitivity and insulin resistance. Serum GLP-1 concentrations were increased significantly to those levels in the control group and correlated with peripheral insulin sensitivity and insulin resistance in patients whose beta-cell functions improved. CONCLUSION Sitagliptin improved beta-cell function, insulin resistance and blood glucose in newly diagnosed patients with T2D. Meanwhile, Sitagliptin ameliorated serum GLP-1 concentrations, which contributed to the enhancement of beta-cell.
Collapse
Affiliation(s)
- Tuan Dinh Le
- Department of Internal Medicine, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | - Nga Thi Phi Nguyen
- Department of Endocrinology, Military Hospital 103, Ha Noi, Vietnam
- Department of Rheumatology and Endocrinology, Vietnam Military Medical University, Ha Noi, Vietnam
| | - Son Tien Nguyen
- Department of Endocrinology, Military Hospital 103, Ha Noi, Vietnam
- Department of Rheumatology and Endocrinology, Vietnam Military Medical University, Ha Noi, Vietnam
| | | | | | - Hoang Huy Duong
- Department of Internal Medicine, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | | | - Binh Nhu Do
- Division of Military Science, Military Hospital 103, Ha Noi, Vietnam
| |
Collapse
|
35
|
Zhang C, Ye F, Wang J, He P, Lei M, Huang L, Huang A, Tang P, Lin H, Liao Y, Liang Y, Ni J, Yan P. Design, Synthesis, and Evaluation of a Series of Novel Super Long-Acting DPP-4 Inhibitors for the Treatment of Type 2 Diabetes. J Med Chem 2020; 63:7108-7126. [PMID: 32452679 DOI: 10.1021/acs.jmedchem.0c00374] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the present work, a novel series of trifluoromethyl-substituted tetrahydropyran derivatives were rationally designed and synthesized as potent DPP-4 inhibitors with significantly improved duration time of action over current commercially available DPP-4 inhibitors. The incorporation of the trifluoromethyl group on the 6-position of the tetrahydropyran ring of omarigliptin with the configuration of (2R,3S,5R,6S) not only significantly improves the overall pharmacokinetic profiles in mice but also maintains comparable DPP-4 inhibition activities. Further preclinical development of compound 2 exhibited its extraordinary efficacy in vivo and good safety profile. Clinical studies of compound 2 (Haisco HSK7653) are now ongoing in China, which revealed that inhibitor 2 could serve as an efficient candidate with a once-biweekly therapeutic regimen.
Collapse
Affiliation(s)
- Chen Zhang
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Fei Ye
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Jianmin Wang
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Ping He
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Ming Lei
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Longbin Huang
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Anbang Huang
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Pingming Tang
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Hongjun Lin
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Yuting Liao
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Yong Liang
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Jia Ni
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| | - Pangke Yan
- Haisco Pharmaceutical Group Company Ltd., 136 Baili Road, Wenjiang district, Chengdu 611130, China
| |
Collapse
|
36
|
Eckhardt BA, Rowsey JL, Thicke BS, Fraser DG, O’Grady KL, Bondar OP, Hines JM, Singh RJ, Thoreson AR, Rakshit K, Lagnado AB, Passos JF, Vella A, Matveyenko AV, Khosla S, Monroe DG, Farr JN. Accelerated osteocyte senescence and skeletal fragility in mice with type 2 diabetes. JCI Insight 2020; 5:135236. [PMID: 32267250 PMCID: PMC7253018 DOI: 10.1172/jci.insight.135236] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
The worldwide prevalence of type 2 diabetes (T2D) is increasing. Despite normal to higher bone density, patients with T2D paradoxically have elevated fracture risk resulting, in part, from poor bone quality. Advanced glycation endproducts (AGEs) and inflammation as a consequence of enhanced receptor for AGE (RAGE) signaling are hypothesized culprits, although the exact mechanisms underlying skeletal dysfunction in T2D are unclear. Lack of inducible models that permit environmental (in obesity) and temporal (after skeletal maturity) control of T2D onset has hampered progress. Here, we show in C57BL/6 mice that a onetime pharmacological intervention (streptozotocin, STZ) initiated in adulthood combined with high-fat diet-induced (HFD-induced) obesity caused hallmark features of human adult-onset T2D, including prolonged hyperglycemia, insulin resistance, and pancreatic β cell dysfunction, but not complete destruction. In addition, HFD/STZ (i.e., T2D) resulted in several changes in bone quality that closely mirror those observed in humans, including compromised bone microarchitecture, reduced biomechanical strength, impaired bone material properties, altered bone turnover, and elevated levels of the AGE CML in bone and blood. Furthermore, T2D led to the premature accumulation of senescent osteocytes with a unique proinflammatory signature. These findings highlight the RAGE pathway and senescent cells as potential targets to treat diabetic skeletal fragility.
Collapse
Affiliation(s)
| | | | | | - Daniel G. Fraser
- Division of Endocrinology
- Robert and Arlene Kogod Center on Aging
| | | | | | | | | | - Andrew R. Thoreson
- Materials and Structural Testing Core
- Department of Physical Medicine and Rehabilitation, and
| | - Kuntol Rakshit
- Division of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Anthony B. Lagnado
- Robert and Arlene Kogod Center on Aging
- Division of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - João F. Passos
- Robert and Arlene Kogod Center on Aging
- Division of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Aleksey V. Matveyenko
- Division of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sundeep Khosla
- Division of Endocrinology
- Robert and Arlene Kogod Center on Aging
| | - David G. Monroe
- Division of Endocrinology
- Robert and Arlene Kogod Center on Aging
| | - Joshua N. Farr
- Division of Endocrinology
- Robert and Arlene Kogod Center on Aging
- Division of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
37
|
Myricitrin Ameliorates Hyperglycemia, Glucose Intolerance, Hepatic Steatosis, and Inflammation in High-Fat Diet/Streptozotocin-Induced Diabetic Mice. Int J Mol Sci 2020; 21:ijms21051870. [PMID: 32182914 PMCID: PMC7084451 DOI: 10.3390/ijms21051870] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023] Open
Abstract
To test the hypothesis that myricitrin (MYR) improves type 2 diabetes, we examined the effect of MYR on hyperglycemia, glucose intolerance, hepatic steatosis, and inflammation in high-fat diet (HFD) and streptozotocin (STZ)-induced type 2 diabetic mice. Male C57BL/6J mice were randomly divided into three groups: non-diabetic, diabetic control, and MYR (0.005%, w/w)-supplemented diabetic groups. Diabetes was induced by HFD and STZ, and MYR was administered orally for 5 weeks. Myricitrin exerted no significant effects on food intake, body weight, fat weight, or plasma lipids levels. However, MYR significantly decreased fasting blood glucose levels, improved glucose intolerance, and increased pancreatic β-cell mass compared to the diabetic control group. Myricitrin administration also markedly increased glucokinase mRNA expression and activity as well as lowered glucose-6-phosphatase and phosphoenolpyruvate carboxykinase mRNA expression and activity in the liver. In addition, liver weight, hepatic triglyceride content, and lipid droplet accumulation were markedly decreased following MYR administration. These changes were seemingly attributable to the suppression of the hepatic lipogenic enzymes—fatty acid synthase and phosphatidate phosphohydrolase. Myricitrin also significantly lowered plasma MCP-1 and TNF-α levels and the mRNA expression of hepatic pro-inflammatory genes. These results suggest that MYR has anti-diabetic potential.
Collapse
|
38
|
Nauck MA. The rollercoaster history of using physiological and pharmacological properties of incretin hormones to develop diabetes medications with a convincing benefit-risk relationship. Metabolism 2020; 103:154031. [PMID: 31785258 DOI: 10.1016/j.metabol.2019.154031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Michael A Nauck
- Diabetes Division, Katholisches Klinikum Bochum, St. Josef Hospital (Ruhr-University Bochum), Gudrunstr. 56, 44791 Bochum, Germany.
| |
Collapse
|
39
|
Onoue T, Goto M, Wada E, Furukawa M, Okuji T, Okada N, Kobayashi T, Iwama S, Sugiyama M, Tsunekawa T, Takagi H, Hagiwara D, Ito Y, Morishita Y, Seino Y, Suga H, Banno R, Hamada Y, Ando M, Yamamori E, Arima H. Dipeptidyl peptidase-4 inhibitor anagliptin reduces fasting apolipoprotein B-48 levels in patients with type 2 diabetes: A randomized controlled trial. PLoS One 2020; 15:e0228004. [PMID: 31990936 PMCID: PMC6986701 DOI: 10.1371/journal.pone.0228004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 01/03/2020] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes and dyslipidemia are diseases that collectively increase the risk of patients developing cardiovascular complications. Several incretin-based drugs are reported to improve lipid metabolism, and one of these medications, anagliptin, is a dipeptidyl peptidase-4 (DPP-4) inhibitor that has been shown to decrease serum triglyceride and low-density lipoproteins cholesterol. This study aimed to conduct an investigation into the effects of anagliptin on serum lipid profiles. This multicenter, open-label, randomized (1:1), parallel group study was designed to evaluate the effects of anagliptin on serum lipid profiles (triglycerides, lipoproteins, apolipoproteins, and cholesterol fractions). The study involved 24 patients with type 2 diabetes at two participating hospitals for a period of 24 weeks. Patients were randomly assigned to the anagliptin (n = 12) or control (n = 12) groups. Patients in the anagliptin group were treated with 200 mg of the drug twice daily. Patients in the control group did not receive anagliptin, but continued with their previous treatment schedules. Lipid metabolism was examined under fasting conditions at baseline and 24 weeks. Patients treated with anagliptin for 24 weeks exhibited significantly reduced levels of serum apolipoprotein B-48, a marker for lipid transport from the intestine, compared with the control group patients (P < 0.05). After 24 weeks of treatment, serum adiponectin levels were significantly raised, whereas glycated hemoglobin (HbA1c) levels were significantly lower compared with the baseline in the anagliptin group (P < 0.05), but not in the control group. This study showed that the DPP-4 inhibitor anagliptin reduces fasting apolipoprotein B-48 levels, suggesting that this drug may have beneficial effects on lipid metabolism possibly mediated by the inhibition of intestinal lipid transport.
Collapse
Affiliation(s)
- Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail: (TOn); (HA)
| | - Motomitsu Goto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Eri Wada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Furukawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayuki Okuji
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Okada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiaki Morishita
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Seino
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryoichi Banno
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, Japan
| | - Yoji Hamada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Ando
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Etsuko Yamamori
- Kainan Hospital, Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Aichi, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail: (TOn); (HA)
| |
Collapse
|
40
|
Abdel-Rahman RF, Ezzat SM, Ogaly HA, Abd-Elsalam RM, Hessin AF, Fekry MI, Mansour DF, Mohamed SO. Ficus deltoidea extract down-regulates protein tyrosine phosphatase 1B expression in a rat model of type 2 diabetes mellitus: a new insight into its antidiabetic mechanism. J Nutr Sci 2020; 9:e2. [PMID: 32042410 PMCID: PMC6984126 DOI: 10.1017/jns.2019.40] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022] Open
Abstract
Ficus deltoidea var. deltoidea Jack (FD) is a well-known plant used in Malay folklore medicine to lower blood glucose in diabetic patients. For further research of the antihyperglycemic mechanisms, the protein tyrosine phosphatase 1B (PTP1B)-inhibitory effect of FD was analysed both in vitro and in vivo. To optimise a method for FD extraction, water, 50, 70, 80, 90 and 95 % ethanol extracts were prepared and determined for their total phenolic and triterpene contents, and PTP1B-inhibition capacity. Among the tested extracts, 70 % ethanol FD extract showed a significant PTP1B inhibition (92·0 % inhibition at 200 µg/ml) and high phenolic and triterpene contents. A bioassay-guided fractionation of the 70 % ethanol extract led to the isolation of a new triterpene (3β,11β-dihydroxyolean-12-en-23-oic acid; F3) along with six known compounds. In vivo, 4 weeks' administration of 70 % ethanol FD extract (125, 250 and 500 mg/kg/d) to streptozotocin-nicotinamide-induced type 2 diabetic rats reversed the abnormal changes of blood glucose, insulin, total Hb, GLUT2, lipid profile, and oxidative stress in liver and pancreas. Moreover, FD reduced the mRNA expression of the key gluconeogenic enzymes (phosphoenolpyruvate carboxykinase and glucose 6-phosphatase) and restored insulin receptor and GLUT2 encoding gene (Slc2a2) expression. In addition, FD significantly down-regulated the hepatic PTP1B gene expression. These results revealed that FD could potentially improve insulin sensitivity, suppress hepatic glucose output and enhance glucose uptake in type 2 diabetes mellitus through down-regulation of PTP1B. Together, our findings give scientific evidence for the traditional use of FD as an antidiabetic agent.
Collapse
Key Words
- CAT, catalase
- Dihydroxyolean-12-en-23-oic acid
- FBG, fasting blood glucose
- FD, Ficus deltoidea var. deltoidea Jack
- Ficus deltoidea
- G6Pase, glucose 6-phosphatase
- GPx, glutathione peroxidase
- GSH, reduced glutathione
- Glucose 6-phosphatase
- Glucose transporter-2
- MDA, malondialdehyde
- MET, metformin
- NA, nicotinamide
- PEPCK, phosphoenolpyruvate carboxykinase
- PTP, protein tyrosine phosphatase
- Phosphoenolpyruvate carboxykinase
- Protein tyrosine phosphatase 1B
- SOD, superoxide dismutase
- STZ, streptozotocin
- Slc2a2, GLUT2 gene
- T2DM, type 2 diabetes mellitus
Collapse
Affiliation(s)
| | - Shahira M. Ezzat
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Einy Street, Cairo11562, Egypt
- Pharmacognosy Department, Faculty of Pharmacy, October University for Modern Sciences and Arts, 6th October Campus, 12566, Egypt
| | - Hanan A. Ogaly
- Chemistry Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Biochemistry Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Reham M. Abd-Elsalam
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Alyaa F. Hessin
- Pharmacology Department, National Research Centre, Giza, Egypt
- Microbiology and Immunology Department, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Mostafa I. Fekry
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Einy Street, Cairo11562, Egypt
| | - Dina F. Mansour
- Pharmacology Department, National Research Centre, Giza, Egypt
| | - Shanaz O. Mohamed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| |
Collapse
|
41
|
Hou Y, Lin S, Qiu J, Sun W, Dong M, Xiang Y, Wang L, Du P. NLRP3 inflammasome negatively regulates podocyte autophagy in diabetic nephropathy. Biochem Biophys Res Commun 2020; 521:791-798. [DOI: 10.1016/j.bbrc.2019.10.194] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 10/29/2019] [Indexed: 01/14/2023]
|
42
|
Shawky LM, Morsi AA, El Bana E, Hanafy SM. The Biological Impacts of Sitagliptin on the Pancreas of a Rat Model of Type 2 Diabetes Mellitus: Drug Interactions with Metformin. BIOLOGY 2019; 9:E6. [PMID: 31881657 PMCID: PMC7167819 DOI: 10.3390/biology9010006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022]
Abstract
Sitagliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor, is a beneficial class of antidiabetic drugs. However, a major debate about the risk of developing pancreatitis is still existing. The aim of the work was to study the histological and immunohistochemical effects of sitagliptin on both endocrine and exocrine pancreases in a rat model of type 2 diabetes mellitus and to correlate these effects with the biochemical findings. Moreover, a possible synergistic effect of sitagliptin, in combination with metformin, was also evaluated. Fifty adult male rats were used and assigned into five equal groups. Group 1 served as control. Group 2 comprised of untreated diabetic rats. Group 3 diabetic rats received sitagliptin. Group 4 diabetic rats received metformin. Group 5 diabetic rats received both combined. Treatments were given for 4 weeks after the induction of diabetes. Blood samples were collected for biochemical assay before the sacrification of rats. Pancreases were removed, weighed, and were processed for histological and immunohistochemical examination. In the untreated diabetic group, the islets appeared shrunken with disturbed architecture and abnormal immunohistochemical reactions for insulin, caspase-3, and inducible nitric oxide synthase (iNOS). The biochemical findings were also disturbed. Morphometrically, there was a significant decrease in the islet size and islet number. Treatment with sitagliptin, metformin, and their combination showed an improvement, with the best response in the combined approach. No evidence of pancreatic injury was identified in the sitagliptin-treated groups. In conclusion, sitagliptin had a cytoprotective effect on beta-cell damage. Furthermore, the data didn't indicate any detrimental effects of sitagliptin on the exocrine pancreas.
Collapse
Affiliation(s)
- Lamiaa M. Shawky
- Department of Histology and Cell Biology, Benha Faculty of Medicine, Benha University, Benha 13511, Egypt;
| | - Ahmed A. Morsi
- Department of Histology and Cell Biology, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt
| | - Eman El Bana
- Department of Anatomy, Benha Faculty of Medicine, Benha University, Benha 13511, Egypt;
| | - Safaa Masoud Hanafy
- Department of Anatomy, Faculty of Medicine for Girls, Al-Azhar University, Cairo 11865, Egypt;
| |
Collapse
|
43
|
Abdel-Kader MS, Soliman GA, Abdel-Rahman RF, Saeedan AS, Abd-Elsalam RM, Ogaly HA. Effect of olive leaves extract on the antidiabetic effect of glyburide for possible herb-drug interaction. Saudi Pharm J 2019; 27:1182-1195. [PMID: 31885478 PMCID: PMC6921200 DOI: 10.1016/j.jsps.2019.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/02/2019] [Indexed: 02/02/2023] Open
Abstract
The concomitant use of olive leaves (OL) and glyburide (GLB) is a possible therapy for diabetic patients. However, there is no report about the effect of OL on the antidiabetic effect of GLB till now. In the current study, the possible interaction of olive leaves extract (OLE) with GLB was assessed to determine if there was any pharmacological benefit over GLB alone. Seven groups of male Sprague Dawley rats were used. Normal rats of the 1st group treated with 2 mL/kg of 3% Tween 80 (vehicle). The 2nd–5th groups were diabetic rats received vehicle, GLB (5 mg/kg), OLE low dose and OLE high dose respectively, while the 6th–7th groups administered combinations of GLB plus OLE low dose and GLB plus OLE high dose, respectively. All treatments were administered orally once daily for 8 weeks. The use of GLB+OLE-500 obviously improved fasting blood glucose (FBG), insulin and glycated hemoglobin (HbA1c) in diabetic rats (95.5 ± 5.55 mg/dL, 6.8 ± 0.16 mg/dL and 6.1 ± 0.29%, respectively) compared to those treated with GLB monotherapy (140.0 ± 6.36 mg/dL, 5.4 ± 0.19 mg/dL and 7.0 ± 0.20%, respectively). The lipid profile [triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C)] was significantly improved in diabetic rats exposed to GLB+OLE-500 (35.6 ± 1.51 mg/dL, 48.5 ± 2.74 mg/dL, 25.1 ± 1.21 mg/dL and 17.0 ± 0.82 mg/dL, respectively) in comparison with diabetic group exposed to GLB alone (43.2 ± 2.15 mg/dL, 56.8 ± 2.14 mg/dL, 18.6 ± 0.96 mg/dL, 23.0 ± 1.26 mg/dL, respectively). Additionally, the benefit impacts of GLB+OLE-500GLB+OLE-500 therapy on the antioxidant and lipid peroxidation parameters in the pancreatic tissues of diabetic rats were higher than those of GLB monotherapy. Moreover, GLB plus OLE-500 combination had the greatest effect on restoration of the insulin content of Beta (β) cells and reduction of the glucagon and somatostatin of Alpha (α) and Delta (δ) endocrine cells in the pancreatic islets among the different treatment. The current study suggests that OL and GLB combination could cause herb-drug interactions through modulation of insulin receptor (INR), glucose transporter 2 (Slc2a2) and peroxisome proliferator-activated receptor α (PPAR-α) genes expression in the liver of diabetic rats.
Collapse
Affiliation(s)
- Maged S Abdel-Kader
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia.,Department of Pharmacognosy, College of Pharmacy, Alexandria University, Alexandria 21215, Egypt
| | - Gamal A Soliman
- Department of Pharmacology, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia.,Department of Pharmacology, College of Veterinary Medicine, Cairo University, Giza, Egypt
| | | | - Abdulaziz S Saeedan
- Department of Pharmacology, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Reham M Abd-Elsalam
- Department of Pathology, College of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hanan A Ogaly
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Biochemistry, College of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
44
|
Liu HT, Gao Y. Efficacy of short-term intensive treatment with insulin pump to improve islet β-cell function in newly diagnosed type 2 diabetes via inhibition of oxidative stress. Exp Ther Med 2019; 18:2293-2298. [PMID: 31452715 DOI: 10.3892/etm.2019.7821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 04/11/2019] [Indexed: 11/06/2022] Open
Abstract
The present study (Chinese Trial Registry GTB7027) assessed the effects of short-term intensive treatment with insulin pump on islet cell function in patients with newly diagnosed type 2 diabetes and the possible mechanism. A total of 100 patients newly diagnosed with type 2 diabetes and hospitalized between January 2016 and December 2017 were divided into a control and an experimental group (n=50 in each group). The subjects of the control group were administered multiple insulin injections for intensive treatment, while the experimental group received short-term intensive treatment with an insulin pump. Analysis of blood parameters, including lipids and glucose, as well as islet β-cell function were performed. The level of reactive oxygen species (ROS) in the peripheral blood mononuclear cells (PBMCs) from the patients was also measured. Oxidative stress indicators, including serum malondialdehyde (MDA) and superoxide dismutase (SOD), were also examined to explore the possible mechanism. The mRNA expression of heme oxygenase 1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) in PBMCs were analyzed by reverse transcription-quantitative PCR. The results indicated that the blood lipid levels were significantly improved in the two groups at two weeks, while the experimental group had significantly lower levels of total cholesterol and triglyceride, as well as low- and high-density lipoprotein cholesterol. The function of islet β-cells was significantly improved in the two groups. The insulin secretion index [homeostasis model assessment (HOMA) of β-cell function] in the experimental group was higher, while the insulin resistance (IR) index (HOMA of IR) was significantly lower than that in the control group. The serum MDA level in the experimental group was significantly lower and the SOD level was significantly higher compared with that in the control group. Following treatment, the level of ROS in diabetic PBMCs was significantly reduced, and the transcription level of HO-1 and Nrf2 were also significantly reduced (P<0.05). These results demonstrated that short-term intensive treatment with an insulin pump significantly improved lipid and blood glucose metabolism to protect islet function as well as significantly reducing the level of oxidative stress in patients with newly diagnosed type 2 diabetes.
Collapse
Affiliation(s)
- Hai-Tong Liu
- Endocrine Department, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163001, P.R. China
| | - Yan Gao
- Endocrine Department, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163001, P.R. China
| |
Collapse
|
45
|
Wang Y, Zhou H, Palyha O, Mu J. Restoration of insulin receptor improves diabetic phenotype in T2DM mice. JCI Insight 2019; 4:124945. [PMID: 31391336 DOI: 10.1172/jci.insight.124945] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 06/27/2019] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM), also known as adult-onset diabetes, is characterized by ineffective insulin action due to insulin resistance in key metabolic tissues. Insulin receptor (IR) plays an important role in insulin signal transduction, defect of which has been considered the fundamental cause of T2DM. IR content reduction in diabetes is one key contributor to the defective insulin signaling and diabetes progression. Rescuing IR levels by transgenic complementation has not been considered as a treatment option because it is limited by uncontrollable expression level, tissue selectivity, or developmental defects. In the current study, we demonstrated that single-dose adeno-associated virus (AAV) vector delivered expression of human IR (hIR) in the liver of inducible IR-knockout mice and significantly improved the diabetic phenotype caused by IR deletion during adulthood. Such an approach was also applied, for the first time to our knowledge, to treating ob/ob mice, a model of severe T2DM attributed to superfluous calorie intake and insulin resistance. Interestingly, similar treatment with AAV-hIR had no obvious effect in healthy animals, indicative of low hypoglycemic risk as a consequence of potential excessive insulin action. The results described here support restoration of IR expression as a safe and effective T2DM therapeutic with a long-lasting profile.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Blood Glucose/analysis
- Dependovirus/genetics
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/therapy
- Disease Models, Animal
- Genetic Therapy/adverse effects
- Genetic Therapy/methods
- Genetic Vectors/administration & dosage
- Genetic Vectors/adverse effects
- Genetic Vectors/genetics
- Humans
- Hypoglycemia/blood
- Hypoglycemia/diagnosis
- Hypoglycemia/genetics
- Insulin/metabolism
- Male
- Mice
- Mice, Knockout
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Treatment Outcome
Collapse
|
46
|
Marques C, Gonçalves A, Pereira PMR, Almeida D, Martins B, Fontes-Ribeiro C, Reis F, Fernandes R. The dipeptidyl peptidase 4 inhibitor sitagliptin improves oxidative stress and ameliorates glomerular lesions in a rat model of type 1 diabetes. Life Sci 2019; 234:116738. [PMID: 31398418 DOI: 10.1016/j.lfs.2019.116738] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/29/2019] [Accepted: 08/05/2019] [Indexed: 12/13/2022]
Abstract
AIMS Oxidative stress has been linked to the development and progression of diabetic nephropathy (DN). The present study evaluated whether the dipeptidyl peptidase-4 inhibitor sitagliptin attenuates glomerular lesions and oxidative stress evoked by chronic hyperglycemia, by a mechanism independent of insulin secretion and glycemia normalization. MAIN METHODS A rat model of DN caused by streptozotocin injection was established and the effects of sitagliptin (5 mg/kg/day) were evaluated after two weeks of treatment. KEY FINDINGS Sitagliptin treatment did not change body weight, glycemic and lipid profiles. However, histopathological observation revealed that sitagliptin attenuates diabetes-induced glomerular lesions on diabetic rats. Sitagliptin also ameliorated the increase in DPP-4 content and promoted the stabilization of GLP-1 in the diabetic kidney. Furthermore, sitagliptin treatment significantly attenuated the increase of free-radical formation and the decrease of antioxidant defenses, attenuating therefore the oxidative stress in the kidneys of diabetic animals. SIGNIFICANCE The results suggest that sitagliptin treatment alleviates kidney oxidative stress in type 1 diabetic rats, which could play a key role in reducing the progression of DN.
Collapse
Affiliation(s)
- Catarina Marques
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Andreia Gonçalves
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Patrícia Manuela Ribeiro Pereira
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniela Almeida
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Beatriz Martins
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Carlos Fontes-Ribeiro
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Flávio Reis
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Rosa Fernandes
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
47
|
Contribution of intestinal dipeptidyl peptidase-4 inhibition for incretin-dependent improved glucose tolerance in mice. Eur J Pharmacol 2019; 859:172521. [PMID: 31276666 DOI: 10.1016/j.ejphar.2019.172521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/21/2019] [Accepted: 07/01/2019] [Indexed: 11/22/2022]
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors prevent the degradation of glucagon-like peptide-1 (GLP-1) and improve glycemic control. The GLP-1 insulinotropic effect involves a pathway through vagus nerve GLP-1 receptors in the gut, in addition to a direct effect on the pancreas. Therefore, this study verified whether DPP-4 inhibition in the gut contributed to the improvement of glycemic control. Anagliptin, a DPP-4 inhibitor, was administered orally or subcutaneously (with or without passing through the gastrointestinal tract, respectively) to mice. The association between blood glucose suppression following oral glucose challenge and DPP-4 inhibition in the small intestine and plasma was assessed. Oral administration of anagliptin (0.03-0.3 mg/kg) in normal mice significantly suppressed blood glucose, which was associated with an increase in insulin secretion at a dose of ≥0.1 mg/kg (P < 0.05). Subcutaneous administration of anagliptin (0.01-0.1 mg/kg) produced similar results. However, plasma DPP-4 inhibition following oral administration was weaker than that following subcutaneous administration; blood glucose suppression was significantly correlated with small intestinal DPP-4 inhibition (r = 0.949, P < 0.01), but not with plasma DPP-4 inhibition. Additionally, similar results were observed in a type 2 diabetes model (r = 0.975, P < 0.001). Thus, these results demonstrated that an improvement in glycemic control was dependent upon small intestinal DPP-4 inhibition. As these effects were accompanied by the elevation of intact GLP-1 in the portal, this suggests that improvement in glucose tolerance after anagliptin treatment might be related to an increase in GLP-1 receptor signaling in the small intestine and portal vein.
Collapse
|
48
|
Böni-Schnetzler M, Häuselmann SP, Dalmas E, Meier DT, Thienel C, Traub S, Schulze F, Steiger L, Dror E, Martin P, Herrera PL, Gabay C, Donath MY. β Cell-Specific Deletion of the IL-1 Receptor Antagonist Impairs β Cell Proliferation and Insulin Secretion. Cell Rep 2019; 22:1774-1786. [PMID: 29444430 DOI: 10.1016/j.celrep.2018.01.063] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 12/18/2017] [Accepted: 01/19/2018] [Indexed: 11/27/2022] Open
Abstract
Interleukin-1 receptor antagonist (IL-1Ra) is elevated in the circulation during obesity and type 2 diabetes (T2D) but is decreased in islets from patients with T2D. The protective role of local IL-1Ra was investigated in pancreatic islet β cell (βIL-1Ra)-specific versus myeloid-cell (myeloIL-1Ra)-specific IL-1Ra knockout (KO) mice. Deletion of IL-1Ra in β cells, but not in myeloid cells, resulted in diminished islet IL-1Ra expression. Myeloid cells were not the main source of circulating IL-1Ra in obesity. βIL-1Ra KO mice had impaired insulin secretion, reduced β cell proliferation, and decreased expression of islet proliferation genes, along with impaired glucose tolerance. The key cell-cycle regulator E2F1 partly reversed IL-1β-mediated inhibition of potassium channel Kir6.2 expression and rescued impaired insulin secretion in IL-1Ra knockout islets. Our findings provide evidence for the importance of β cell-derived IL-1Ra for the local defense of β cells to maintain normal function and proliferation.
Collapse
Affiliation(s)
- Marianne Böni-Schnetzler
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland.
| | - Stéphanie P Häuselmann
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Elise Dalmas
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Daniel T Meier
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Constanze Thienel
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Shuyang Traub
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Friederike Schulze
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Laura Steiger
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Erez Dror
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Praxedis Martin
- Department of Pathology and Immunology, Centre Médical Universitaire, 1211 Geneva 4, Switzerland
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211 Geneva, Switzerland; Centre Facultaire du Diabète, University of Geneva, 1211 Geneva, Switzerland
| | - Cem Gabay
- Department of Pathology and Immunology, Centre Médical Universitaire, 1211 Geneva 4, Switzerland
| | - Marc Y Donath
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
49
|
Small molecule inhibition of dipeptidyl peptidase-4 enhances bone marrow progenitor cell function and angiogenesis in diabetic wounds. Transl Res 2019; 205:51-63. [PMID: 30452888 PMCID: PMC7252504 DOI: 10.1016/j.trsl.2018.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 09/29/2018] [Accepted: 10/25/2018] [Indexed: 12/23/2022]
Abstract
In diabetes, stromal cell-derived factor-1 (SDF-1) expression and progenitor cell recruitment are reduced. Dipeptidyl peptidase-4 (DPP-4) inhibits SDF-1 expression and progenitor cell recruitment. Here we examined the impact of the DPP-4 inhibitor, MK0626, on progenitor cell kinetics in the context of wound healing. Wildtype (WT) murine fibroblasts cultured under high-glucose to reproduce a diabetic microenvironment were exposed to MK0626, glipizide, or no treatment, and SDF-1 expression was measured with ELISA. Diabetic mice received MK0626, glipizide, or no treatment for 6 weeks and then were wounded. Immunohistochemistry was used to quantify neovascularization and SDF-1 expression. Gene expression was measured at the RNA and protein level using quantitative polymerase chain reaction and ELISA, respectively. Flow cytometry was used to characterize bone marrow-derived mesenchymal progenitor cell (BM-MPC) population recruitment to wounds. BM-MPC gene expression was assayed using microfluidic single cell analysis. WT murine fibroblasts exposed to MK0626 demonstrated increased SDF-1 expression. MK0626 treatment significantly accelerated wound healing and increased wound vascularity, SDF-1 expression, and dermal thickness in diabetic wounds. MK0626 treatment increased the number of BM-MPCs present in bone marrow and in diabetic wounds. MK0626 had no effect on BM-MPC population dynamics. BM-MPCs harvested from MK0626-treated mice exhibited increased chemotaxis in response to SDF-1 when compared to diabetic controls. Treatment with a DPP-4 inhibitor significantly improved wound healing, angiogenesis, and endogenous progenitor cell recruitment in the setting of diabetes.
Collapse
|
50
|
Chemoprotective effects of butanol fraction of Buchholzia coriacea (Capparidaceae) against type 2 diabetes and oxidative stress in male Wistar rats. Biosci Rep 2019; 39:BSR20170665. [PMID: 28790167 PMCID: PMC6379225 DOI: 10.1042/bsr20170665] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/26/2017] [Accepted: 08/02/2017] [Indexed: 12/29/2022] Open
Abstract
Recent studies have shown that Type 2 diabetes (T2D) in rats can result through a synergy that links obesity to insulin resistance and β-cell dysfunction. The present study achieved T2D via high fructose (20%w/v, p.o.), streptozotocin single dose (40 mg/kg, i.p.) (HFSTZ) in rats. Also, chemoprotective potential of butanol fraction of Buchholzia coriacea (BFBC) was demonstrated. Control normal and diabetic untreated (HFSTZ-induced T2D) rats received CM-cellulose (1 mg/kg, p.o.). Diabetic rats received intragastric BFBC (20, 200, 400 mg/kg), glibenclamide (0.07 mg/kg), and BFBC (200 mg/kg) plus glibenclamide treatments, respectively. 2,2-Diphenyl-1-picrylhydrazyl, nitric oxide radical, hydroxyl radical scavenging activities, and α-amylase inhibition were assessed. After 2 weeks of treatments, blood glucose levels, lipid profiles, renal and liver function, serum insulin as well as in vivo oxidative stress biomarkers were assessed. BFBC shows highest antioxidants and α-amylase inhibitory activities in vitro HFSTZ-induced T2D produced hyperglycemia (P<0.05-0.001; F = 5.26-26.47), serum hyperinsulinemia (six-folds) plus elevated lipid peroxidation levels. Similarly, there were altered lipid profiles, liver and renal biomarker enzymes plus weight loss. BFBC administration alone or in combination with glibenclamide reversed T2D symptomatologies in treated animals, and improved body weights against control diabetic rats. In vivo antioxidant activities also improved while histological sections in treated rats show reduced tissue damage in pancreas, kidneys, liver, and heart, respectively. Oleic, stearic, 2-methyl-pyrrolidine-2-carboxylic, and n-hexadecanoic acids were present in BFBC in large quantities given GC-MS analysis. Overall, data from the present study suggest chemoprotective potentials of BFBC against HFSTZ-induced T2D rats.
Collapse
|