1
|
Lim CG, Ozkan B, Liang Y, Chen J, Yao J, Khaing NEE, Rooney MR, Ndumele CE, Tai ES, Coresh J, Sim X, van Dam RM. Plasma Proteomic Signatures of Adiposity Are Associated With Cardiovascular Risk Factors and Type 2 Diabetes Risk in a Multiethnic Asian Population. Diabetes 2025; 74:416-426. [PMID: 39621883 PMCID: PMC11842604 DOI: 10.2337/db24-0184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 11/25/2024] [Indexed: 02/22/2025]
Abstract
The biomarkers connecting obesity and cardiometabolic diseases are not fully understood. We aimed to 1) evaluate the associations between BMI, waist circumference (WC), and ∼5,000 plasma proteins (SomaScan V4), 2) identify protein signatures of BMI and WC, and 3) evaluate the associations between the protein signatures and cardiometabolic health, including metabolically unhealthy obesity and type 2 diabetes incidence in the Singapore Multi-Ethnic Cohort Phase 1 (MEC1). Among 410 BMI-associated and 385 WC-associated proteins, we identified protein signatures of BMI and WC and validated them in an independent data set across two time points and externally in the Atherosclerosis Risk in Communities (ARIC) study. The BMI and WC protein signatures were highly correlated with total and visceral body fat, respectively. Furthermore, the protein signatures were significantly associated with cardiometabolic risk factors and metabolically unhealthy obesity. In prospective analyses, the protein signatures were strongly associated with type 2 diabetes risk in MEC1 (odds ratio per SD increment in WC protein signature 2.84; 95% CI 2.47-3.25) and ARIC (hazard ratio 1.98; 95% CI 1.88-2.08). Our protein signatures have potential uses in the monitoring of metabolically unhealthy obesity. ARTICLE HIGHLIGHTS We evaluated the associations between ∼5,000 plasma proteins and BMI and waist circumference (WC) in a multiethnic Asian population. We identified 410 proteins associated with BMI and 385 proteins associated with WC and derived protein signatures of BMI and WC, which we validated externally in a U.S. cohort. Both the BMI and WC protein signatures were strongly associated with cardiometabolic risk factors, metabolically unhealthy obesity, and risk of obesity, metabolic syndrome, and type 2 diabetes. Our protein signatures have potential uses in monitoring metabolically unhealthy obesity.
Collapse
Affiliation(s)
- Charlie G.Y. Lim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Bige Ozkan
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Yujian Liang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Jingsha Chen
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Jiali Yao
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Nang Ei Ei Khaing
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Mary R. Rooney
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Chiadi E. Ndumele
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Josef Coresh
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Rob M. van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Departments of Exercise and Nutrition Sciences and Epidemiology, Milken Institute School of Public Health, George Washington University, Washington, DC
| |
Collapse
|
2
|
Li P, He X, Feng E, Wei J, Tu H, Chen T. Lactobacillus acidophilus JYLA-126 Ameliorates Obesity-Associated Metabolic Disorders by Positively Regulating the AMPK Signaling Pathway Through the Gut-Liver Axis. Probiotics Antimicrob Proteins 2025; 17:62-80. [PMID: 38051435 DOI: 10.1007/s12602-023-10190-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
Obesity is a chronic metabolic disease worldwide and is considered a major health problem in contemporary society. Lactobacillus acidophilus have demonstrated beneficial effects on obesity, but the specific mechanism of how it exerts beneficial effects has not been elucidated. Here, we found that L. acidophilus JYLA-126 had good biological properties for intestinal health, such as antioxidation, acid tolerance, bile salt tolerance, antimicrobial activity, and gut colonization. We further identified that supplementation of L. acidophilus JYLA-126 obese mice possessed a dose-dependent amelioration of body weight, intestinal imbalance, and metabolic disorders compared to HFD-induced mice. Mechanistically, the excellent slimming effect of L. acidophilus JYLA-126 was achieved mainly by reversing HFD-induced gut dysbiosis, inhibiting inflammatory factors and balancing the homeostasis of the gut-liver axis. Specifically, L. acidophilus JYLA-126 improved hepatic glycogen synthesis, lowered oxidative stress, and facilitated lipid metabolism by regulating AMPK signaling pathway-related protein expression to restore the overall metabolic level. Accordingly, L. acidophilus JYLA-126 promoted energy uptake efficiency in obese mice, resulting in significant expression of uncoupling protein 1 (UCP1) protein in brown adipose tissue (BAT), and markedly reduced the size of adipocytes. These findings indicate that the anti-obesity activity of L. acidophilus JYLA-126 correlates with activation of the AMPK signaling pathway through improved gut-liver interactions.
Collapse
Affiliation(s)
- Ping Li
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Xia He
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Enxu Feng
- College of Food Science and Engineering, Shandong Agricultural University, Taian, 271018, People's Republic of China
| | - Jing Wei
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Huaijun Tu
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China.
| | - Tingtao Chen
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China.
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China.
| |
Collapse
|
3
|
Jiang L, Huang L, Dai C, Zheng R, Miyake M, Mori Y, Nakao S, Morino K, Ymashiro K, Miao Y, Li Q, Ren W, Ye Z, Li H, Yang Z, Shi Y. Genome-Wide Association Analysis Identifies LILRB2 Gene for Pathological Myopia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308968. [PMID: 39207058 PMCID: PMC11516067 DOI: 10.1002/advs.202308968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/13/2024] [Indexed: 09/04/2024]
Abstract
Pathological myopia (PM) is one of the leading causes of blindness, especially in Asia. To identify the genetic risk factors of PM, a two-stage genome-wide association study (GWAS) and replication analysis in East Asian populations is conducted. The analysis identified LILRB2 in 19q13.42 as a new candidate locus for PM. The increased protein expression of LILRB2/Pirb (mouse orthologous protein) in PM patients and myopia mouse models is validated. It is further revealed that the increase in LILRB2/Pirb promoted fatty acid synthesis and lipid accumulation, leading to the destruction of choroidal function and the development of PM. This study revealed the association between LILRB2 and PM, uncovering the molecular mechanism of lipid metabolism disorders leading to the pathogenesis of PM due to LILRB2 upregulation.
Collapse
Affiliation(s)
- Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Chao Dai
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Rui Zheng
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Masahiro Miyake
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Yuki Mori
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Shin‐ya Nakao
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Kazuya Morino
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Kenji Ymashiro
- Department of Ophthalmology and Visual SciencesKyoto University Graduate School of MedicineKyoto606‐8501Japan
| | - Yang‐Bao Miao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Qi Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Weiming Ren
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Zimeng Ye
- School of MedicineUniversity of SydneyCamperdownNSW2050Australia
| | - Hongjing Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
- Jinfeng Laboratory, Chongqing, ChinaChongqing400000China
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduSichuan610072China
| |
Collapse
|
4
|
Gong K, Xue C, Feng Z, Pan R, Wang M, Chen S, Chen Y, Guan Y, Dai L, Zhang S, Jiang L, Li L, Wang B, Yin Z, Ma L, Iwakiri Y, Tang J, Liao C, Chen H, Duan Y. Intestinal Nogo-B reduces GLP1 levels by binding to proglucagon on the endoplasmic reticulum to inhibit PCSK1 cleavage. Nat Commun 2024; 15:6845. [PMID: 39122737 PMCID: PMC11315690 DOI: 10.1038/s41467-024-51352-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Glucagon-like peptide 1 (GLP1), which is mainly processed and cleaved from proglucagon in enteroendocrine cells (EECs) of the intestinal tract, acts on the GLP1 receptor in pancreatic cells to stimulate insulin secretion and to inhibit glucagon secretion. However, GLP1 processing is not fully understood. Here, we show that reticulon 4B (Nogo-B), an endoplasmic reticulum (ER)-resident protein, interacts with the major proglucagon fragment of proglucagon to retain proglucagon on the ER, thereby inhibiting PCSK1-mediated cleavage of proglucagon in the Golgi. Intestinal Nogo-B knockout in male type 2 diabetes mellitus (T2DM) mice increases GLP1 and insulin levels and decreases glucagon levels, thereby alleviating pancreatic injury and insulin resistance. Finally, we identify aberrantly elevated Nogo-B expression and inhibited proglucagon cleavage in EECs from diabetic patients. Our study reveals the subcellular regulatory processes involving Nogo-B during GLP1 production and suggests intestinal Nogo-B as a potential therapeutic target for T2DM.
Collapse
Affiliation(s)
- Ke Gong
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui, China
| | - Chao Xue
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Zian Feng
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ruru Pan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Mengyao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui, China
| | - Shasha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui, China
| | - Yudong Guan
- Department of Geriatrics, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lingyun Dai
- Department of Geriatrics, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui, China
| | - Liwei Jiang
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Ling Li
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Bei Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Zequn Yin
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Likun Ma
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Junming Tang
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui, China
| | - Houzao Chen
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
5
|
Wang T, Zhang M, Gong X, Chen W, Peng Y, Liao C, Xu H, Li Q, Shen G, Ren H, Zhu Y, Zhang B, Mao J, Wei L, Chen Y, Yang X. Inhibition of Nogo-B reduces the progression of pancreatic cancer by regulation NF-κB/GLUT1 and SREBP1 pathways. iScience 2024; 27:109741. [PMID: 38706871 PMCID: PMC11068639 DOI: 10.1016/j.isci.2024.109741] [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: 10/10/2023] [Revised: 01/22/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024] Open
Abstract
Pancreatic cancer (PC) is a lethal disease and associated with metabolism dysregulation. Nogo-B is related to multiple metabolic related diseases and types of cancers. However, the role of Nogo-B in PC remains unknown. In vitro, we showed that cell viability and migration was largely reduced in Nogo-B knockout or knockdown cells, while enhanced by Nogo-B overexpression. Consistently, orthotopic tumor and metastasis was reduced in global Nogo knockout mice. Furthermore, we indicated that glucose enhanced cell proliferation was associated to the elevation expression of Nogo-B and nuclear factor κB (NF-κB). While, NF-κB, glucose transporter type 1 (GLUT1) and sterol regulatory element-binding protein 1 (SREBP1) expression was reduced in Nogo-B deficiency cells. In addition, we showed that GLUT1 and SREBP1 was downstream target of NF-κB. Therefore, we demonstrated that Nogo deficiency inhibited PC progression is regulated by the NF-κB/GLUT1 and SREBP1 pathways, and suggested that Nogo-B may be a target for PC therapy.
Collapse
Affiliation(s)
- Tianxiang Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Min Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Xinyu Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Wanjing Chen
- Department of General Surgery, The Second Affiliated Hospital, Anhui Medical University, Hefei 230000, China
| | - Ying Peng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Hongmei Xu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Qingshan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Guodong Shen
- Department of Geriatrics, The First Affiliated Hospital of University of Science and Technology of China, Gerontology Institute of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Huirong Ren
- Department of Geriatrics, The First Affiliated Hospital of University of Science and Technology of China, Gerontology Institute of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Yaxin Zhu
- Institute for International Health Professions Education and Research, China Medical University, Shenyang 110000, China
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
| | - Jiali Mao
- Department of Anesthesiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230000, China
| | - Lingling Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| |
Collapse
|
6
|
Li J, Zhang S, Sun Y, Li J, Feng Z, Li H, Zhang M, Yan T, Han J, Duan Y. Liver ChREBP deficiency inhibits fructose-induced insulin resistance in pregnant mice and female offspring. EMBO Rep 2024; 25:2097-2117. [PMID: 38532128 PMCID: PMC11014959 DOI: 10.1038/s44319-024-00121-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
High fructose intake during pregnancy increases insulin resistance (IR) and gestational diabetes mellitus (GDM) risk. IR during pregnancy primarily results from elevated hormone levels. We aim to determine the role of liver carbohydrate response element binding protein (ChREBP) in insulin sensitivity and lipid metabolism in pregnant mice and their offspring. Pregnant C57BL/6J wild-type mice and hepatocyte-specific ChREBP-deficient mice were fed with a high-fructose diet (HFrD) or normal chow diet (NC) pre-delivery. We found that the combination of HFrD with pregnancy excessively activates hepatic ChREBP, stimulating progesterone synthesis by increasing MTTP expression, which exacerbates IR. Increased progesterone levels upregulated hepatic ChREBP via the progesterone-PPARγ axis. Placental progesterone activated the progesterone-ChREBP loop in female offspring, contributing to IR and lipid accumulation. In normal dietary conditions, hepatic ChREBP modestly affected progesterone production and influenced IR during pregnancy. Our findings reveal the role of hepatic ChREBP in regulating insulin sensitivity and lipid homeostasis in both pregnant mice consuming an HFrD and female offspring, and suggest it as a potential target for managing gestational metabolic disorders, including GDM.
Collapse
Affiliation(s)
- Jiaqi Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuyao Sun
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Jian Li
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300052, China
| | - Zian Feng
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huaxin Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mengxue Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tengteng Yan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.
| | - Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
7
|
Gong K, Zhang Z, Chen SS, Zhu XR, Wang MY, Yang XY, Ding C, Han JH, Li QS, Duan YJ. 6-Methyl flavone inhibits Nogo-B expression and improves high fructose diet-induced liver injury in mice. Acta Pharmacol Sin 2023; 44:2216-2229. [PMID: 37402997 PMCID: PMC10618526 DOI: 10.1038/s41401-023-01121-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
Excessive fructose consumption increases hepatic de novo lipogenesis, resulting in cellular stress, inflammation and liver injury. Nogo-B is a resident protein of the endoplasmic reticulum that regulates its structure and function. Hepatic Nogo-B is a key protein in glycolipid metabolism, and inhibition of Nogo-B has protective effects against metabolic syndrome, thus small molecules that inhibit Nogo-B have therapeutic benefits for glycolipid metabolism disorders. In this study we tested 14 flavones/isoflavones in hepatocytes using dual luciferase reporter system based on the Nogo-B transcriptional response system, and found that 6-methyl flavone (6-MF) exerted the strongest inhibition on Nogo-B expression in hepatocytes with an IC50 value of 15.85 μM. Administration of 6-MF (50 mg· kg-1 ·d-1, i.g. for 3 weeks) significantly improved insulin resistance along with ameliorated liver injury and hypertriglyceridemia in high fructose diet-fed mice. In HepG2 cells cultured in a media containing an FA-fructose mixture, 6-MF (15 μM) significantly inhibited lipid synthesis, oxidative stress and inflammatory responses. Furthermore, we revealed that 6-MF inhibited Nogo-B/ChREBP-mediated fatty acid synthesis and reduced lipid accumulation in hepatocytes by restoring cellular autophagy and promoting fatty acid oxidation via the AMPKα-mTOR pathway. Thus, 6-MF may serve as a potential Nogo-B inhibitor to treat metabolic syndrome caused by glycolipid metabolism dysregulation.
Collapse
Affiliation(s)
- Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Zhen Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Sha-Sha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Xin-Ran Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Meng-Yao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Xin-Yue Yang
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Chen Ding
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Ji-Hong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qing-Shan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China.
| | - Ya-Jun Duan
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
8
|
Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, Tian Y, Fu X. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (Beijing) 2023; 4:e283. [PMID: 37303813 PMCID: PMC10248034 DOI: 10.1002/mco2.283] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents one of the fastest growing epidemic metabolic disorders worldwide and is a strong contributor for a broad range of comorbidities, including vascular, visual, neurological, kidney, and liver diseases. Moreover, recent data suggest a mutual interplay between T2DM and Corona Virus Disease 2019 (COVID-19). T2DM is characterized by insulin resistance (IR) and pancreatic β cell dysfunction. Pioneering discoveries throughout the past few decades have established notable links between signaling pathways and T2DM pathogenesis and therapy. Importantly, a number of signaling pathways substantially control the advancement of core pathological changes in T2DM, including IR and β cell dysfunction, as well as additional pathogenic disturbances. Accordingly, an improved understanding of these signaling pathways sheds light on tractable targets and strategies for developing and repurposing critical therapies to treat T2DM and its complications. In this review, we provide a brief overview of the history of T2DM and signaling pathways, and offer a systematic update on the role and mechanism of key signaling pathways underlying the onset, development, and progression of T2DM. In this content, we also summarize current therapeutic drugs/agents associated with signaling pathways for the treatment of T2DM and its complications, and discuss some implications and directions to the future of this field.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Huimin Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yu Zhang
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Geng Liu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Haixia Xu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Guocheng Rao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
9
|
Zheng J, Wang S, Zhang T, Li H, Zhu M, Wei X, Ge Y, Yang X, Zhang S, Xu H, Duan Y, Liu L, Chen Y. Nogo-B inhibition restricts ulcerative colitis via inhibiting p68/miR-155 signaling pathway. Int Immunopharmacol 2023; 120:110378. [PMID: 37244119 DOI: 10.1016/j.intimp.2023.110378] [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: 03/28/2023] [Revised: 05/10/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
BACKGROUND & AIMS Ulcerative colitis (UC) is a main type of inflammatory bowel diseases which spreads globally during the westernization of lifestyle over the past few decades. However, the cause of UC is still not fully understood. We aimed to disclose the role of Nogo-B in the development of UC. METHODS Nogo-deficiency (Nogo-/-) and wild-type male mice were treated with dextran sodium sulfate (DSS) to conduct a UC model, followed by determination of colon and serum inflammatory cytokines level. RAW264.7, THP1 and NCM460 cells were used to determine macrophage inflammation as well as proliferation and migration of NCM460 cells under Nogo-B or miR-155 intervention. RESULTS Nogo deficiency significantly reduced DSS-induced weight loss, colon length and weight reduction, and inflammatory cells accumulation in the intestinal villus, while increased the expression of tight junctions (TJs) proteins (Zonula occludens-1, Occludin) and adherent junctions (AJs) proteins (E-cadherin, α-catenin), implying that Nogo deficiency attenuated DSS-induced UC. Mechanistically, Nogo-B deficiency reduced TNFα, IL-1β and IL-6 levels in the colon, serum, RAW264.7 cells and THP1-derived macrophages. Furthermore, we identified that Nogo-B inhibition can reduce the maturation of miR-155, which is essential for Nogo-B-affected inflammatory cytokines expression. Interestingly, we determined that Nogo-B and p68 can interact with each other to promote the expression and activation of Nogo-B and p68, thus facilitating miR-155 maturation to induce macrophage inflammation. Blocking p68 inhibited Nogo-B, miR-155, TNFα, IL-1β and IL-6 expression. Moreover, the culture medium collected from Nogo-B overexpressed macrophages can inhibit enterocytes NCM460 cells proliferation and migration. CONCLUSION We disclose that Nogo deficiency reduced DSS-induced UC via inhibiting p68-miR-155-activated inflammation. Our results indicate that Nogo-B inhibition serves as a new potential therapeutic candidate for the prevention and treatment of UC.
Collapse
Affiliation(s)
- Juan Zheng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shengnan Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tingting Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Huaxin Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mengmeng Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoning Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yu Ge
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Hongmei Xu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lipei Liu
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
10
|
Du K, Chen GH, Bai X, Chen L, Hu SQ, Li YH, Wang GZ, He JW, Lai SJ. Dynamics of transcriptome and chromatin accessibility revealed sequential regulation of potential transcription factors during the brown adipose tissue whitening in rabbits. Front Cell Dev Biol 2022; 10:981661. [PMID: 36225319 PMCID: PMC9548568 DOI: 10.3389/fcell.2022.981661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Brown adipose tissue (BAT) represents a valuable target for treating obesity in humans. BAT losses of thermogenic capacity and gains a “white adipose tissue-like (WAT-like)” phenotype (BAT whitening) under thermoneutral environments, which could lead to potential low therapy responsiveness in BAT-based obesity treatments. However, the epigenetic mechanisms of BAT whitening remain largely unknown. In this study, BATs were collected from rabbits at day0 (D0), D15, D85, and 2 years (Y2). RNA-sequencing (RNA-seq) and the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) were performed to investigate transcriptome and chromatin accessibility of BATs at the four whitening stages, respectively. Our data showed that many genes and chromatin accessible regions (refer to as “peaks”) were identified as significantly changed during BAT whitening in rabbits. The BAT-selective genes downregulated while WAT-selective genes upregulated from D0 to Y2, and the de novo lipogenesis-related genes reached the highest expression levels at D85. Both the highly expressed genes and accessible regions in Y2 were significantly enriched in immune response-related signal pathways. Analysis of different relationships between peaks and their nearby genes found an increased proportion of the synchronous changes between chromatin accessibility and gene expression during BAT whitening. The synergistic changes between the chromatin accessibility of promoter and the gene expression were found in the key adipose genes. The upregulated genes which contained increased peaks were significantly enriched in the PI3K-Akt signaling pathway, steroid biosynthesis, TGF-beta signaling pathway, osteoclast differentiation, and dilated cardiomyopathy. Moreover, the footprinting analysis suggested that sequential regulation of potential transcription factors (TFs) mediated the loss of thermogenic phenotype and the gain of a WAT-like phenotype of BAT. In conclusion, our study provided the transcriptional and epigenetic frameworks for understanding BAT whitening in rabbits for the first time and might facilitate potential insights into BAT-based obesity treatments.
Collapse
Affiliation(s)
- Kun Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Guan-He Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xue Bai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Li Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shen-Qiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yan-Hong Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Guo-Ze Wang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jing-Wei He
- Sichuan Animal Husbandry Station, Chengdu, China
| | - Song-Jia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Song-Jia Lai,
| |
Collapse
|
11
|
Wolosowicz M, Prokopiuk S, Kaminski TW. Recent Advances in the Treatment of Insulin Resistance Targeting Molecular and Metabolic Pathways: Fighting a Losing Battle? MEDICINA (KAUNAS, LITHUANIA) 2022; 58:472. [PMID: 35454311 PMCID: PMC9029454 DOI: 10.3390/medicina58040472] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022]
Abstract
Diabetes Mellitus (DM) is amongst the most notable causes of years of life lost worldwide and its prevalence increases perpetually. The disease is characterized as multisystemic dysfunctions attributed to hyperglycemia resulting directly from insulin resistance (IR), inadequate insulin secretion, or enormous glucagon secretion. Insulin is a highly anabolic peptide hormone that regulates blood glucose levels by hastening cellular glucose uptake as well as controlling carbohydrate, protein, and lipid metabolism. In the course of Type 2 Diabetes Mellitus (T2DM), which accounts for nearly 90% of all cases of diabetes, the insulin response is inadequate, and this condition is defined as Insulin Resistance. IR sequela include, but are not limited to, hyperglycemia, cardiovascular system impairment, chronic inflammation, disbalance in oxidative stress status, and metabolic syndrome occurrence. Despite the substantial progress in understanding the molecular and metabolic pathways accounting for injurious effects of IR towards multiple body organs, IR still is recognized as a ferocious enigma. The number of widely available therapeutic approaches is growing, however, the demand for precise, safe, and effective therapy is also increasing. A literature search was carried out using the MEDLINE/PubMed, Google Scholar, SCOPUS and Clinical Trials Registry databases with a combination of keywords and MeSH terms, and papers published from February 2021 to March 2022 were selected as recently published papers. This review paper aims to provide critical, concise, but comprehensive insights into the advances in the treatment of IR that were achieved in the last months.
Collapse
Affiliation(s)
- Marta Wolosowicz
- Department of Physiology, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Slawomir Prokopiuk
- Faculty of Health Sciences, Lomza State University of Applied Sciences, 18-400 Lomza, Poland;
| | - Tomasz W. Kaminski
- Department of Medicine, Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|