1
|
Jahdkaran M, Sistanizad M. From lipids to glucose: Investigating the role of dyslipidemia in the risk of insulin resistance. J Steroid Biochem Mol Biol 2025; 250:106744. [PMID: 40158704 DOI: 10.1016/j.jsbmb.2025.106744] [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: 01/18/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Dyslipidemia is recognized as one of the most prevalent metabolic disorders and is frequently associated with other prevalent conditions, particularly diabetes mellitus. There appears to be a bidirectional connection between these two metabolic disorders. While considerable research has focused on how insulin resistance can lead to lipid abnormalities, the reverse relationship specifically, how dyslipidemia could assist in developing insulin resistance and diabetes mellitus has received relatively less attention. This review aims to comprehensively evaluate the mechanisms through which dyslipidemia can induce insulin resistance. Dyslipidemia is primarily classified into three main categories: hypercholesterolemia, hypertriglyceridemia, and low levels of HDL. These conditions may promote insulin resistance across multiple pathways, including the accumulation of lipid metabolites, dysfunction of pancreatic β-cells, increased reactive oxygen species, endoplasmic reticulum stress and inflammation, endothelial dysfunction, alterations in adiponectin levels, changes in bile acid composition and concentration, and dysbiosis of gut microbiota. However, further investigation is required to fully elucidate the cellular and molecular mechanisms underlying the relationship between lipid disorders and insulin resistance. Emphasizing such research could facilitate the development of therapeutic strategies targeting both conditions simultaneously.
Collapse
Affiliation(s)
- Mahtab Jahdkaran
- Prevention of Cardiovascular Disease Research Center, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sistanizad
- Prevention of Cardiovascular Disease Research Center, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Commissati S, Cagigas ML, Masedunskas A, Petrucci G, Tosti V, De Ciutiis I, Rajakumar G, Kirmess KM, Meyer MR, Goldhamer A, Kennedy BK, Hatem D, Rocca B, Fiorito G, Fontana L. Prolonged fasting promotes systemic inflammation and platelet activation in humans: A medically supervised, water-only fasting and refeeding study. Mol Metab 2025; 96:102152. [PMID: 40268190 DOI: 10.1016/j.molmet.2025.102152] [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: 03/09/2025] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE Prolonged fasting (PF), defined as abstaining from energy intake for ≥4 consecutive days, has gained interest as a potential health intervention. However, the biological effects of PF on the plasma proteome are not well understood. METHODS In this study, we investigated the effects of a medically supervised water-only fast (mean duration: 9.8 ± 3.1 days), followed by 5.3 ± 2.4 days of guided refeeding, in 20 middle-aged volunteers (mean age: 52.2 ± 11.8 years; BMI: 28.8 ± 6.4 kg/m2). RESULTS Fasting resulted in a 7.7% mean weight loss and significant increases in serum beta-hydroxybutyrate (BHB), confirming adherence. Untargeted high-dimensional plasma proteomics (SOMAScan, 1,317 proteins) revealed multiple adaptations to PF, including preservation of skeletal muscle and bone, enhanced lysosomal biogenesis, increased lipid metabolism via PPARα signaling, and reduced amyloid fiber formation. Notably, PF significantly reduced circulating amyloid beta proteins Aβ40 and Aβ42, key components of brain amyloid plaques. In addition, PF induced an acute inflammatory response, characterized by elevated plasma C-reactive protein (CRP), hepcidin, midkine, and interleukin 8 (IL-8), among others. A retrospective cohort analysis of 1,422 individuals undergoing modified fasting confirmed increased CRP levels (from 2.8 ± 0.1 to 4.3 ± 0.2 mg/L). The acute phase response, associated with transforming growth factor (TGF)-β signaling, was accompanied by increased platelet degranulation and upregulation of the complement and coagulation cascade, validated by ELISAs in blood and urine. CONCLUSIONS While the acute inflammatory response during PF may serve as a transient adaptive mechanism, it raises concerns regarding potential cardiometabolic effects that could persist after refeeding. Further investigation is warranted to elucidate the long-term molecular and clinical implications of PF across diverse populations.
Collapse
Affiliation(s)
| | - Maria Lastra Cagigas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Giovanna Petrucci
- Section of Pharmacology, Department of Safety and Bioethics, Catholic University School of Medicine, Rome, Italy
| | - Valeria Tosti
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Isabella De Ciutiis
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Gayathiri Rajakumar
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore; Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Duaa Hatem
- Section of Pharmacology, Department of Safety and Bioethics, Catholic University School of Medicine, Rome, Italy
| | - Bianca Rocca
- Section of Pharmacology, Department of Safety and Bioethics, Catholic University School of Medicine, Rome, Italy; NeuroFarBa Department, University of Florence, Florence, Italy
| | - Giovanni Fiorito
- Clinical Bioinformatics unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy; MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia.
| |
Collapse
|
3
|
Tao Z, Luo Z, Zou Z, Ye W, Hao Y, Li X, Zheng K, Wu J, Xia J, Zhao Y, Wang Y, Zhang X. Novel insights and an updated review of metabolic syndrome in immune-mediated organ transplant rejection. Front Immunol 2025; 16:1580369. [PMID: 40330480 PMCID: PMC12052740 DOI: 10.3389/fimmu.2025.1580369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic syndrome (MetS) is a group of symptoms that are characterized by abnormal changes in metabolic substances such as glucose, lipids, proteins, and bile acids. MetS is a common complication after organ transplantation and can further affect the survival and physiological function of the graft by reprograming the patient's immune environment. Additionally, MetS can influence the occurrence of post-transplant complications, such as infections. In recent years, research into the epidemiology and mechanisms of MetS has grown significantly. In this review, we summarize the mechanisms of MetS after transplantation and the mechanisms of hyperglycemia, insulin resistance, hyperlipidemia, abnormal bile acids, and abnormal amino acids on the body's immune cells as related to the effect of metabolic disorders on immune rejection after liver, kidney, heart, skin and other organ transplantation. Finally, we provide an overview of current treatment strategies and offer insights into potential future therapies for managing MetS in transplant recipients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yang Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Yongjun Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Accili D, Deng Z, Liu Q. Insulin resistance in type 2 diabetes mellitus. Nat Rev Endocrinol 2025:10.1038/s41574-025-01114-y. [PMID: 40247011 DOI: 10.1038/s41574-025-01114-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 04/19/2025]
Abstract
Insulin resistance is an integral pathophysiological feature of type 2 diabetes mellitus. Here, we review established and emerging cellular mechanisms of insulin resistance, their complex integrative features and their relevance to disease progression. While recognizing the heterogeneity of the elusive fundamental disruptions that cause insulin resistance, we endorse the view that effector mechanisms impinge on insulin receptor signalling and its relationship with plasma levels of insulin. We focus on hyperinsulinaemia and its consequences: acutely impaired but persistent insulin action, with reduced ability to lower glucose levels but preserved lipid synthesis and lipoprotein secretion. We emphasize the role of insulin sensitization as a therapeutic goal in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Domenico Accili
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA.
| | - Zhaobing Deng
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Qingli Liu
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| |
Collapse
|
5
|
Wang S, Qin L, Liu F, Zhang Z. Unveiling the crossroads of STING signaling pathway and metabolic reprogramming: the multifaceted role of the STING in the TME and new prospects in cancer therapies. Cell Commun Signal 2025; 23:171. [PMID: 40197235 PMCID: PMC11977922 DOI: 10.1186/s12964-025-02169-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025] Open
Abstract
The cGAS-STING signaling pathway serves as a critical link between DNA sensing and innate immunity, and has tremendous potential to improve anti-tumor immunity by generating type I interferons. However, STING agonists have shown decreasing biotherapeutic efficacy in clinical trials. Tumor metabolism, characterized by aberrant nutrient utilization and energy production, is a fundamental hallmark of tumorigenesis. And modulating metabolic pathways in tumor cells has been discovered as a therapeutic strategy for tumors. As research concerning STING progressed, emerging evidence highlights its role in metabolic reprogramming, independent its immune function, indicating metabolic targets as a strategy for STING activation in cancers. In this review, we delve into the interplay between STING and multiple metabolic pathways. We also synthesize current knowledge on the antitumor functions of STING, and the metabolic targets within the tumor microenvironment (TME) that could be exploited for STING activation. This review highlights the necessity for future research to dissect the complex metabolic interactions with STING in various cancer types, emphasizing the potential for personalized therapeutic strategies based on metabolic profiling.
Collapse
Affiliation(s)
- Siwei Wang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lu Qin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology), Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhanguo Zhang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
6
|
Dahiya M, Yadav M, Goyal C, Kumar A. Insulin resistance in Alzheimer's disease: signalling mechanisms and therapeutics strategies. Inflammopharmacology 2025; 33:1817-1831. [PMID: 40064805 DOI: 10.1007/s10787-025-01704-2] [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] [Received: 01/26/2025] [Accepted: 02/14/2025] [Indexed: 04/13/2025]
Abstract
BACKGROUND Alzheimer's disease (AD), one of the most common neurodegenerative disorders, is characterised by hallmark abnormalities such as amyloid-β plaques and neurofibrillary tangles (NFTs). Emerging evidence suggests that faulty insulin signalling contributes to these pathological features, impairing critical cellular and metabolic processes. OBJECTIVE This review aims to elucidate the role of insulin signalling in the central nervous system (CNS) under normal and pathological conditions and to explore therapeutic approaches targeting insulin pathways in AD and other neurodegenerative diseases. METHODS We reviewed studies highlighting the involvement of insulin-signalling pathways in neuronal health, with a particular focus on the key components-IRS, PI3K, Akt, and GSK-3β-predominantly expressed in cortical and hippocampal regions. RESULTS Insulin, an essential growth factor, regulates numerous cellular functions, including glucose metabolism, mitochondrial activity, oxidative stress response, autophagy, synaptic plasticity, and cognitive processes. Altered phosphorylation of signalling molecules in insulin pathways contributes to oxidative stress, inflammation, and the formation of AD hallmarks. Indirect modulators such as NF-κB and caspases further exacerbate neuronal damage, linking impaired insulin signalling to neurodegeneration. CONCLUSION Insulin signalling plays a crucial role in maintaining neuronal health and mitigating neurodegenerative processes. Targeting insulin pathways and associated molecules offers promising therapeutic avenues for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Mini Dahiya
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Monu Yadav
- Amity Institute of Pharmacy, Amity University, Haryana, Amity Education Valley Gurugram, Manesar, Panchgaon, Haryana, India
| | - Chetan Goyal
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
7
|
Kim SR, Kim YJ, Kim H, Park S, Jung UJ. Therapeutic Potential of Myricitrin in a db/ db Mouse Model of Type 2 Diabetes. Molecules 2025; 30:1460. [PMID: 40286055 PMCID: PMC11990901 DOI: 10.3390/molecules30071460] [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: 02/25/2025] [Revised: 03/14/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Type 2 diabetes is characterized by insulin resistance, which contributes to dysregulated glucose and lipid metabolism and is associated with chronic inflammation. While previous studies have examined the effects of myricitrin in streptozotocin-induced diabetic models, its impact on the db/db mouse, a model that better reflects insulin resistance-associated metabolic disturbances, remains unclear. In this study, mice were divided into three groups (db/+, db/db, and db/db + 0.02% myricitrin) and were fed their respective diets for five weeks. Myricitrin supplementation reduced fat mass, adipocyte size, and plasma leptin levels, which were elevated in db/db mice. Although myricitrin did not affect fasting blood glucose levels, it lowered plasma insulin, hemoglobin A1c, postprandial glucose levels, and the homeostasis model assessment of insulin resistance, suggesting improvements in insulin sensitivity and glucose homeostasis. Enhanced pancreatic insulin expression, along with reduced hepatic gluconeogenic enzyme activities and mRNA expression, contributed to the improved glucose homeostasis observed in myricitrin-supplemented mice. Additionally, myricitrin reduced hepatic triglyceride levels and lipid droplet accumulation by inhibiting hepatic fatty acid synthase activity. It also decreased plasma inflammatory marker levels and their mRNA expression in adipose tissue. These findings suggest that myricitrin may be a promising therapeutic candidate for type 2 diabetes.
Collapse
Affiliation(s)
- Sang Ryong Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea;
| | - Young-Je Kim
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea;
| | - HwiCheol Kim
- Department of Food Science and Nutrition, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan 48513, Republic of Korea; (H.K.); (S.P.)
| | - Sojeong Park
- Department of Food Science and Nutrition, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan 48513, Republic of Korea; (H.K.); (S.P.)
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan 48513, Republic of Korea; (H.K.); (S.P.)
| |
Collapse
|
8
|
Hung YT, Wu WT, Lee RP, Yao TK, Yeh KT. Beyond Bone Remodeling: Denosumab's Multisystemic Benefits in Musculoskeletal Health, Metabolism, and Age-Related Diseases-A Narrative Review. Biomedicines 2025; 13:732. [PMID: 40149708 PMCID: PMC11940544 DOI: 10.3390/biomedicines13030732] [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: 01/27/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Denosumab, a receptor activator of nuclear factor kappa-Β ligand (RANKL) inhibitor, demonstrates therapeutic effects beyond traditional osteoporosis management through the RANK/RANKL/osteoprotegerin pathway. Methods: This narrative review analyzed 37 studies (2018-2024) examining denosumab's broader physiological effects and clinical applications. Results: Long-term safety data spanning 10 years showed sustained fracture prevention efficacy with a favorable benefit/risk profile. Compared to bisphosphonates, denosumab demonstrated superior outcomes in bone mineral density improvement and fracture risk reduction, particularly in elderly and frail populations. It enhanced muscular function by improving appendicular lean mass and grip strength while reducing fall risk. The drug showed potential cardiovascular benefits through its effects on cardiac and smooth muscle function. Notably, denosumab use was associated with reduced Type II diabetes mellitus risk through improved glucose metabolism. Additionally, it demonstrated promise in osteoarthritis treatment by suppressing osteoclast activity and chondrocyte apoptosis. While there are multisystem benefits, vigilance is required regarding adverse events, including hypocalcemia, infection risk, cutaneous reactions, and osteonecrosis of the jaw. Conclusions: Denosumab exhibits potential benefits in bone and systemic metabolism. Further research is needed to fully understand its therapeutic potential beyond osteoporosis and optimize clinical applications across different populations.
Collapse
Affiliation(s)
- Yi-Ting Hung
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan; (Y.-T.H.); (W.-T.W.)
| | - Wen-Tien Wu
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan; (Y.-T.H.); (W.-T.W.)
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan;
| | - Ru-Ping Lee
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan;
| | - Ting-Kuo Yao
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
| | - Kuang-Ting Yeh
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan; (Y.-T.H.); (W.-T.W.)
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan;
- Graduate Institute of Clinical Pharmacy, Tzu Chi University, Hualien 970374, Taiwan
- Clinical Education, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan
| |
Collapse
|
9
|
Soltanieh SK, Khastar S, Kaur I, Kumar A, Bansal J, Fateh A, Nathiya D, Husseen B, Rajabivahid M, Dehghani-Ghorbi M, Akhavan-Sigari R. Long Non-Coding RNAs in Non-Alcoholic Fatty Liver Disease; Friends or Foes? Cell Biochem Biophys 2025; 83:279-294. [PMID: 39377981 DOI: 10.1007/s12013-024-01555-8] [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: 09/17/2024] [Indexed: 01/03/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a range of conditions that start with the accumulation of fat in the liver (hepatic steatosis) and can progress to more severe stages like steatohepatitis (NASH) and fibrosis without drinking alcohol. Environmental and genetic variables both contribute to MAFLD's development, with various biological processes and mediators involved at every phase. Long non-coding RNAs (lncRNAs) are a class of RNA molecules that are not translated into protein and are over 200 nucleotides long. They can impact genes that encode protein by controlling transcriptional and post-transcriptional procedures. Dysregulation of lncRNA has been connected to several liver diseases, including MAFLD. Recent research has linked lncRNAs to MAFLD pathology in both patients and animal models. However, the roles of most lncRNAs in MAFLD pathology are still not well recognized. This review provides a comprehensive catalog of recently reported lncRNAs in the pathogenesis of MAFLD and summarizes the current knowledge of lncRNAs usage as therapeutic strategies in MAFLD, the most common liver disease. Collectively, lncRNA's targeting could potentially offer a therapeutic approach by modulating MAFLD.
Collapse
Affiliation(s)
| | - Sahar Khastar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka-560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand-831001, India
| | - Jaya Bansal
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, 140307, Punjab, India
| | - Ata Fateh
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| | - Mansour Rajabivahid
- Department of Internal Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warszawa, Poland
| |
Collapse
|
10
|
Lin YK, Pan YF, Jiang TY, Chen YB, Shang TY, Xu MY, Feng HB, Ma YH, Tan YX, Wang HY, Dong LW. Blocking the SIRPα-CD47 axis promotes macrophage phagocytosis of exosomes derived from visceral adipose tissue and improves inflammation and metabolism in mice. J Biomed Sci 2025; 32:31. [PMID: 40016734 PMCID: PMC11869713 DOI: 10.1186/s12929-025-01124-y] [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] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Adipose tissue plays a pivotal role in systemic metabolism and maintaining bodily homeostasis. Exosomes from adipose tissues, known as AT-Exos, are recognized as important messengers in the communication between adipose tissue and other organs. Despite this, the alterations in exosome composition and the functional disparities among depot-specific AT-Exos in obesity remain elusive. METHODS In this work, we utilized lipidomics and microRNA (miRNA) sequencing to elucidate the lipid and miRNA profiles of AT-Exos in a diet-induced obesity model. We identified obesity-related miRNAs in AT-Exos and further explored their mechanisms using gain- and loss-of-function experiments. To evaluate the metabolic effects of AT-Exos on adipocytes, we conducted RNA-sequencing (RNA-seq) and confirmed our findings through Quantitative Real-time PCR (qPCR) and Western bolt analyses. Meanwhile, a mouse model with intraperitoneal injections was utilized to validate the role of exosomes derived from visceral white adipose tissue (vWAT-Exos) in obesity progression in vivo. Finally, we explored potential therapeutic intervention strategies targeting AT-Exos, particularly focusing on modulating the SIRPα-CD47 axis to enhance macrophage phagocytosis using Leptin-deficient (ob/ob) mice and SIRPα knock-out mice. RESULTS Our study revealed that obesity-related metabolism affects the biological processes of AT-Exos, with depot-specific secretion patterns. In obesity, the lipidome profile of AT-Exos was significantly altered, and diet can modify the miRNA content and function within these exosomes, influencing lipid metabolism and inflammatory pathways that contribute to metabolic dysregulation. Specifically, we identified that miR-200a-3p and miR-200b-3p promoted lipid accumulation in 3T3L1 cells partly through the PI3K/AKT/mTOR pathway. RNA-Seq analysis revealed that AT-Exos from different fat depots exerted distinct effects on adipocyte metabolism, with obese vWAT-Exos being notably potent in triggering inflammation and lipid accumulation in diet-induced obesity. Additionally, we found that inhibiting the SIRPα-CD47 axis can mitigate metabolic disorders induced by obese vWAT-Exos or ob/ob mice, partly due to the enhanced clearance of vWAT-Exos. Consistent with this, SIRPα-deficient mice exhibited a reduction in vWAT-Exos and displayed greater resistance to obesity. CONCLUSIONS This study elucidates that diet-induced obesity altered the lipid and miRNA profiles of AT-Exos, which involved in modulating adipocyte inflammation and metabolic balance. The SIRPα-CD47 axis emerges as a potential therapeutic target for obesity and its associated complications.
Collapse
Affiliation(s)
- Yun-Kai Lin
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yu-Fei Pan
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Tian-Yi Jiang
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yi-Bin Chen
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Tai-Yu Shang
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Meng-You Xu
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Hui-Bo Feng
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Yun-Han Ma
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ye-Xiong Tan
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China
| | - Hong-Yang Wang
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Li-Wei Dong
- International Cooperation Laboratory On Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
- Oncology Pharmacy Laboratory, National Center for Liver Cancer, Shanghai, China.
| |
Collapse
|
11
|
Li P, Zhang R, Hu P, Wang T, Wang J. Cepharanthine relieves nonalcoholic steatohepatitis through inhibiting STAT1/CXCL10 axis-mediated lipogenesis and inflammatory responses. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119358. [PMID: 39805479 DOI: 10.1016/j.jep.2025.119358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/24/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Stephania rotunda Lour., a medicinal herb, has been utilized in both Traditional Chinese Medicine (TCM) and Traditional Indian Medicine to treat conditions such as fever, dysentery, and inflammation. Cepharanthine (CEP), a primary active ingredient of Stephania rotunda Lour., has demonstrated a range of pharmacological activities, including anti-oxidative, anti-inflammatory, anti-cancer, anti-viral and anti-parasitic properties. However, the effects and underlying mechanisms of CEP on improving nonalcoholic steatohepatitis (NASH) remain unclear. AIM OF THE STUDY This study aimed to investigate the effects of CEP on mitigating diet-induced NASH and explore its underlying mechanisms. MATERIALS AND METHODS A High-Fat Diet (HFD) and the high levels of free fatty acids (FFA) were used to establish in vivo and in vitro NASH models to evaluate the intervention effect of CEP. Subsequently, RNA-sequencing, western blotting, quantitative real-time PCR (qRT-PCR) and siRNA transfection were employed to investigate its underlying mechanisms. RESULTS Our findings indicated that CEP significantly reduced lipogenesis and inflammatory responses in both HFD-fed rats and FFA-induced hepatic cells (including HepG2, L02 and AML12 cell lines), as is evidenced by the reduction of triglyceride (TG), lipid accumulation, and the release of inflammatory cytokines such as TNF-α, IL-6 and IL-1β. Mechanistically, CEP significantly inhibits CXC motif chemokine ligand 10 (CXCL10) expression both in vivo and in vitro. It also regulates sterol regulatory element binding protein-1c (SREBP1c)-induced lipogenic gene expression and CXCL10-mediated nuclear factor kappa B (NFκB) activation. Notably, knockdown of CXCL10 mimics the ability of CEP to reduce lipid accumulation and inflammatory responses, which is also observed following the blockade of signal transducer and activator of transcription 1 (STAT1) in HepG2 cells. CONCLUSION CEP alleviates NASH by inhibiting lipogenesis and inflammatory responses in a STAT1/CXCL10 axis-dependent manner.
Collapse
Affiliation(s)
- Pan Li
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Ruoyu Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, 999077, PR China
| | - Pingping Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing, 400016, PR China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
12
|
Sedighi M, Saber A, Bagheri A, Hazratian S, Pasdar Y, Najafi F, Moradinazar M, Soleimani D. The associations between diet-induced inflammation and the improvement or worsening of hepatic steatosis and fibrosis: a longitudinal analysis of RaNCD cohort study. Nutr Metab (Lond) 2025; 22:5. [PMID: 39833863 PMCID: PMC11749311 DOI: 10.1186/s12986-025-00897-1] [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: 10/18/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Persistent inflammation plays a crucial role in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). We aimed to scrutinize the associations of diet-induced inflammation with the improvement or worsening of hepatic steatosis and fibrosis in MASLD. METHODS This longitudinal study involved 2,537 participants from the Ravanser Non-Communicable Disease (RaNCD) cohort (2015-2023). Dietary intake was assessed using the 118-item food frequency questionnaire (FFQ), and diet-induced inflammation was determined using the dietary inflammatory index (DII). The AST to platelet ratio index (APRI) and Fibrosis-4 (FIB-4) were used as confirmed predictive indicators for hepatic fibrosis and the hepatic steatosis index (HSI) was used for hepatic steatosis. RESULTS Adherence to an inflammatory diet independently increases the risk of worsening hepatic steatosis (RR:1.39; 95%CI: 1.02-1.93; P-value: 0.04) and reduces the risk of improving hepatic steatosis (RR: 66; 95% CI: 0.48-0.98; P-value: 0.01) compared to an anti-inflammatory diet. The DII scores did not show any connection to hepatic fibrosis, as determined by FIB-4 (β: - 1.08; 95%CI: - 2.43 to 0.27; P-value: 0.12) and APRI (β: 0.22; 95%CI: - 1.51 to 1.95; P-value: 0.80). CONCLUSIONS These results underscore the importance of dietary composition in managing hepatic steatosis and highlight the need for further research to explore the mechanisms underlying these associations.
Collapse
Affiliation(s)
- Mohammad Sedighi
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Dolat-Abad Street, Isar Square, Kermanshah, Iran
| | - Amir Saber
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Dolat-Abad Street, Isar Square, Kermanshah, Iran
| | - Amir Bagheri
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Dolat-Abad Street, Isar Square, Kermanshah, Iran
| | - Saba Hazratian
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Dolat-Abad Street, Isar Square, Kermanshah, Iran
| | - Yahya Pasdar
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farid Najafi
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Moradinazar
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Davood Soleimani
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Dolat-Abad Street, Isar Square, Kermanshah, Iran.
| |
Collapse
|
13
|
Bernecker M, Lin A, Feuchtinger A, Molenaar A, Schriever SC, Pfluger PT. Weight cycling exacerbates glucose intolerance and hepatic triglyceride storage in mice with a history of chronic high fat diet exposure. J Transl Med 2025; 23:7. [PMID: 39754229 PMCID: PMC11699648 DOI: 10.1186/s12967-024-06039-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Obese subjects undergoing weight loss often fear the Yoyo dieting effect, which involves regaining or even surpassing their initial weight. To date, our understanding of such long-term obesity and weight cycling effects is still limited and often based on only short-term murine weight gain and loss studies. This study aimed to investigate the long-term impacts of weight cycling on glycemic control and metabolic health, focusing on adipose tissue, liver, and hypothalamus. METHODS Chow-fed mice and mice subjected to prolonged high-fat diet (HFD) consumption for 20 weeks, followed by 24 weeks of dietary interventions to either induce weight gain, weight loss, or weight cycling were monitored for perturbations in feeding efficiency and glucose homeostasis. Post-mortem analyses included qPCR, Western Blotting, biochemical and microscopical assessments for hepatic steatosis and insulin resistance, hypothalamic and adipose tissue inflammation, and circulating lipid, leptin and IL-6 levels. RESULTS Weight cycling led to hyperphagia and rapid weight regain, matching the weights of mice continuously on HFD. Despite weight loss, adipose tissue inflammation persisted with elevated pro-inflammatory markers, macrophage infiltration, and impaired Glut4 expression. HFD-induced dysregulation in hypothalamic expression of orexigenic peptides and synaptic plasticity markers persisted also after weight normalization suggesting long-lasting neural alterations. Weight-cycled mice exhibited higher circulating IL-6 and leptin levels, increased hepatic lipid storage, and dysregulated glucose metabolism compared to those with consistent diets, indicating worsened metabolic effects by Yoyo dieting. CONCLUSION In sum, our study highlights significant metabolic risks associated with weight cycling, particularly following prolonged obesity. Persistent adipose tissue inflammation, perturbed neural peptide and plasticity markers and impaired glucose tolerance emphasize the need for effective and sustainable weight loss strategies to mitigate the adverse outcomes of weight regain and improve long-term metabolic health.
Collapse
Affiliation(s)
- Miriam Bernecker
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Anna Lin
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology and Tissue Analytics, Helmholtz Munich, Neuherberg, Germany
| | - Anna Molenaar
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Sonja C Schriever
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Paul T Pfluger
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany.
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| |
Collapse
|
14
|
Fraga CG, Cremonini E, Galleano M, Oteiza PI. Natural Products and Diabetes: (-)-Epicatechin and Mechanisms Involved in the Regulation of Insulin Sensitivity. Handb Exp Pharmacol 2025; 287:159-173. [PMID: 38421444 DOI: 10.1007/164_2024_707] [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] [Indexed: 03/02/2024]
Abstract
Type 2 diabetes (T2D) is a disease that occurs when cells do not respond normally to insulin, a condition called insulin resistance, which leads to high blood glucose levels. Although it can be treated pharmacologically, dietary habits beyond carbohydrate restriction can be highly relevant in the management of T2D. Emerging evidence supports the possibility that natural products (NPs) could contribute to managing blood glucose or counteract the undesirable effects of hyperglycemia and insulin resistance. This chapter summarizes the relevant preclinical evidence involving the flavonoid (-)-epicatechin (EC) in the optimization of glucose homeostasis, reducing insulin resistance and/or diabetes-associated disorders. Major effects of EC are observed on (i) intestinal functions, including digestive enzymes, glucose transporters, microbiota, and intestinal permeability, and (ii) redox homeostasis, including oxidative stress and inflammation. There is still a need for further clinical studies to confirm the in vitro and rodent data, allowing recommendations for EC, particularly in prediabetic and T2D patients. The collection of similar data and the lack of clinical evidence for EC is also applicable to other NPs.
Collapse
Affiliation(s)
- Cesar G Fraga
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina.
- Department of Nutrition, University of California, Davis, CA, USA.
| | | | - Monica Galleano
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA, USA
- Department of Environmental Toxicology, University of California, Davis, CA, USA
| |
Collapse
|
15
|
Den Hartogh DJ, MacPherson REK, Tsiani E. Muscle cell palmitate-induced insulin resistance, JNK, IKK/NF-κB, and STAT3 activation are attenuated by carnosic and rosmarinic acid. Appl Physiol Nutr Metab 2025; 50:1-14. [PMID: 39805098 DOI: 10.1139/apnm-2024-0302] [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] [Indexed: 01/16/2025]
Abstract
The worldwide epidemic of obesity has drastically worsened with the increase in more sedentary lifestyles and increased consumption of fatty foods. Increased blood free fatty acids, often observed in obesity, lead to impaired insulin action, and promote the development of insulin resistance and type 2 diabetes mellitus. c-Jun N-terminal kinase (JNK), inhibitor of kappa B (IκB) kinase (IKK)-nuclear factor-kappa B (NF-κB), and signal transducer and activator of transcription 3 (STAT3) are known to be involved in skeletal muscle insulin resistance. We reported previously that carnosic acid (CA) and rosmarinic acid (RA) attenuated the palmitate-induced skeletal muscle insulin resistance, an effect that was associated with increased AMPK activation and reduced mammalian target of rapamycin-p70S6K signaling. In the present study, we examined the effects of CA and RA on JNK, IKK-NF-κB, and STAT3. Exposure of cells to palmitate increased the phosphorylation/activation of JNK, IKKα/β, IκBα, NF-κBp65, and STAT3. Importantly, CA and RA attenuated the deleterious effects of palmitate. Our data indicate that CA and RA have the potential to counteract the palmitate-induced skeletal muscle cell insulin resistance by modulating JNK, IKK-NF-κB, and STAT3 signaling.
Collapse
Affiliation(s)
- Danja J Den Hartogh
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Neuroscience, Brock University, St. Catharines, ON L2S3A1, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
16
|
Pérez-Mingan GC, Sierra-Merlano RM, Yepes I, Vergara MJP, Ortiz M, Peña B, Cano-Pérez E, Gómez-Camargo D. Relationship of Interleukin 6 with Hepatic Steatosis and Liver Fibrosis in Rheumatoid Arthritis at a Rheumatology Care Center in Cartagena, Colombia. Genes (Basel) 2024; 15:1639. [PMID: 39766906 PMCID: PMC11675702 DOI: 10.3390/genes15121639] [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: 11/22/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to investigate the association of IL-6 with steatotic liver disease (SLD) and liver fibrosis (LF) in rheumatoid arthritis (RA) patients at a rheumatology center in Cartagena de Indias, Colombia. METHODS This was a cross-sectional study that included RA and non-RA cases. The level of cellular expression of interleukin 6 (IL-6) was evaluated by flow cytometry in peripheral blood leukocytes, and the presence of SLD and LF was detected by elastosonography. The main outcome was to establish the association between the levels of cellular expression of IL-6 and the development of SLD and LF. RESULTS This study included 47 cases of RA and 34 cases on-RA, with a mean age of 54 and 55 years, respectively. The frequency of SLD was 55.3% in RA and 38.2% in non-RA. The frequency of LF was 12.8% in RA and 14.7% in non-RA, with no statistical difference. The levels of cellular expression of IL-6 were significantly higher in RA compared to non-RA. Cellular expression of IL-6 was associated with the presence of SLD (54% vs. 30.3%; p = 0.002). This association was not maintained in RA cases (49.5% vs. 47.6%; p = 0.571). No association was found between cellular expression of IL-6 and LF in the total population (43.8% vs. 42.7%; p = 0.813) nor in RA cases (59.41% vs. 48.3%; p = 0.526). CONCLUSIONS IL-6 levels were related to SLD in the evaluated sample, and RA was not a risk factor for SLD or LF. The prognostic role of IL-6 for SLD in patients with RA requires further studies.
Collapse
Affiliation(s)
- Gloria Caterine Pérez-Mingan
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia; (G.C.P.-M.); (R.M.S.-M.); (I.Y.)
| | - Rita Magola Sierra-Merlano
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia; (G.C.P.-M.); (R.M.S.-M.); (I.Y.)
| | - Ismael Yepes
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia; (G.C.P.-M.); (R.M.S.-M.); (I.Y.)
| | | | - Miguel Ortiz
- Programa de Medicina, Facultad de Ciencias de la Salud, Universidad del Sinú, Cartagena 130001, Colombia;
| | - Breiner Peña
- Programa de Medicina, Facultad de Ciencias de la Salud, Universidad del Magdalena, Santa Marta 470001, Colombia;
| | - Eder Cano-Pérez
- Grupo de Investigación UNIMOL, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia;
- Doctorado en Medicina Tropical, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia
| | - Doris Gómez-Camargo
- Grupo de Investigación UNIMOL, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia;
- Doctorado en Medicina Tropical, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia
| |
Collapse
|
17
|
Lin Z, Lin J, Huang A, Zhang Z, Wu X, Yin G, Wei C, Xu W. Angiotensin (1-7) Improves Pancreatic Islet Function via Upregulating PDX-1 and GCK: A Dose-Dependent Study in Mice. Int J Endocrinol 2024; 2024:1672096. [PMID: 39734383 PMCID: PMC11671625 DOI: 10.1155/ije/1672096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose: This study aimed to verify the effect of angiotensin (1-7) on improving islet function and further explore the signaling pathway that may be involved in this improvement. It also aimed to explore the effects of angiotensin (1-7) on blood glucose levels, islet function, and morphological changes in db/db mice and its potential signal pathway. Methods: Forty-five db/db mice were divided randomly into a model control group and different doses of angiotensin (1-7) intervention groups (0, 150, 300, and 600 μg/kg/d), while seven db/m mice were assigned as the normal control group. The angiotensin (1-7) intervention groups received daily intraperitoneal administration for 8 weeks, whereas the normal control group was injected intraperitoneally with an equal volume of normal saline every day for 8 weeks. Changes in weight and food intake of mice were detected. Effect of angiotensin (1-7) on lipid metabolism, islet function, the morphology of pancreatic islets, and β-cell mass on mice were evaluated. The expression of PDX-1 and GCK in pancreatic tissue was verified. Results: The group receiving angiotensin (1-7) at a dosage of 600 μg/kg/d showed a significant decrease in body weight, triglyceride levels, and fasting blood glucose, along with an improvement in glucose tolerance. In the 300 μg/kg/d group, angiotensin (1-7) tended to increase the total volume of islets. Moreover, the intervention groups exhibited a significant increase in the ratio of β cells, small islets (30-80 μm in diameter), as well as the expression levels of PDX-1 and GCK in pancreatic tissue. Conclusion: Angiotensin (1-7) could improve glucose and lipid metabolism and islet function by promoting the expression of PDX-1 and GCK genes in the pancreas of db/db mice.
Collapse
Affiliation(s)
- Ziwei Lin
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jiaqi Lin
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Anqi Huang
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zixu Zhang
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xinyi Wu
- Shantou University Medical College, Shantou, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guoshu Yin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chiju Wei
- Multidisciplinary Research Center, Shantou University, Shantou, China
| | - Wencan Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
18
|
Li L, Cai F, Guo C, Liu Z, Qin J, Huang J. Gut microbiome and NAFLD: impact and therapeutic potential. Front Microbiol 2024; 15:1500453. [PMID: 39664063 PMCID: PMC11632136 DOI: 10.3389/fmicb.2024.1500453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/13/2024] [Indexed: 12/13/2024] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) affects approximately 32.4% of the global population and poses a significant health concern. Emerging evidence underscores the pivotal role of the gut microbiota-including bacteria, viruses, fungi, and parasites-in the development and progression of NAFLD. Dysbiosis among gut bacteria alters key biological pathways that contribute to liver fat accumulation and inflammation. The gut virome, comprising bacteriophages and eukaryotic viruses, significantly shapes microbial community dynamics and impacts host metabolism through complex interactions. Similarly, gut fungi maintain a symbiotic relationship with bacteria; the relationship between gut fungi and bacteria is crucial for overall host health, with certain fungal species such as Candida in NAFLD patients showing detrimental associations with metabolic markers and liver function. Additionally, the "hygiene hypothesis" suggests that reduced exposure to gut parasites may affect immune regulation and metabolic processes, potentially influencing conditions like obesity and insulin resistance. This review synthesizes current knowledge on the intricate interactions within the gut microbiota and their associations with NAFLD. We highlight the therapeutic potential of targeting these microbial communities through interventions such as probiotics, prebiotics, and fecal microbiota transplantation. Addressing the complexities of NAFLD requires comprehensive strategies that consider the multifaceted roles of gut microorganisms in disease pathology.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
19
|
Zhong K, Du X, Niu Y, Li Z, Tao Y, Wu Y, Zhang R, Guo L, Bi Y, Tang L, Dou T, Wang L. Progress in the mechanism of functional dyspepsia: roles of mitochondrial autophagy in duodenal abnormalities. Front Med (Lausanne) 2024; 11:1491009. [PMID: 39655235 PMCID: PMC11627220 DOI: 10.3389/fmed.2024.1491009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Mitochondria are the main source of energy for cellular activity. Their functional damage or deficiency leads to cellular deterioration, which in turn triggers autophagic reactions. Taking mitochondrial autophagy as a starting point, the present review explored the mechanisms of duodenal abnormalities in detail, including mucosal barrier damage, release of inflammatory factors, and disruption of intracellular signal transduction. We summarized the key roles of mitochondrial autophagy in the abnormal development of the duodenum and examined the in-depth physiological and pathological mechanisms involved, providing a comprehensive theoretical basis for understanding the pathogenesis of functional dyspepsia. At present, it has been confirmed that an increase in the eosinophil count and mast cell degranulation in the duodenum can trigger visceral hypersensitive reactions and cause gastrointestinal motility disorders. In the future, it is necessary to continue exploring the molecular mechanisms and signaling pathways of mitochondrial autophagy in duodenal abnormalities. A deeper understanding of mitochondrial autophagy provides important references for developing treatment strategies for functional dyspepsia, thereby improving clinical efficacy and patient quality of life.
Collapse
Affiliation(s)
- Kexin Zhong
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaojuan Du
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yuanyuan Niu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhengju Li
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yongbiao Tao
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yuqian Wu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Ruiting Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linjing Guo
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yurong Bi
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lijuan Tang
- College of Pharmacy, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tianyu Dou
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Longde Wang
- Department of Gastroenterology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
20
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
21
|
Jiang F, Wang L, Ying H, Sun J, Zhao J, Lu Y, Bian Z, Chen J, Fang A, Zhang X, Larsson SC, Mantzoros CS, Wang W, Yuan S, Ding Y, Li X. Multisystem health comorbidity networks of metabolic dysfunction-associated steatotic liver disease. MED 2024; 5:1413-1423.e3. [PMID: 39116870 DOI: 10.1016/j.medj.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/09/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND The global burden of metabolic dysfunction-associated steatotic liver disease (MASLD) is growing, but its subsequent health consequences have not been thoroughly examined. METHODS A phenome-wide association study was conducted to map the associations of MASLD with 948 unique clinical outcomes among 361,021 Europeans in the UK Biobank. Disease trajectory and comorbidity analyses were applied to visualize the sequential patterns of multiple comorbidities related to the occurrence of MASLD. The associations jointly verified by observational and polygenic phenome-wide analyses were further replicated by two-sample Mendelian randomization analysis using data from the FinnGen study and international consortia. FINDINGS The observational and polygenic phenome-wide association study revealed the associations of MASLD with 96 intrahepatic and extrahepatic diseases, including circulatory, metabolic, genitourinary, neurological, gastrointestinal, and hematologic diseases. Sequential patterns of MASLD-related extrahepatic comorbidities were primarily found in circulatory, metabolic, and inflammatory diseases. Mendelian randomization analyses supported the causal associations between MASLD and the risk of several intrahepatic disorders, metabolic diseases, cardio-cerebrovascular disease, and ascites but found no associations with neurological diseases. CONCLUSIONS This study elucidated multisystem comorbidities and health consequences of MASLD, contributing to the development of combination interventions targeting distinct pathways for health promotion among patients with MASLD. FUNDING X.L. was funded by the Natural Science Fund for Distinguished Young Scholars of Zhejiang Province (LR22H260001) and the National Nature Science Foundation of China (82204019) and Y.D. was funded by the Key Project of Traditional Chinese Medicine Science and Technology Plan of Zhejiang Province (GZY-ZJ-KJ-24077) and the National Natural Science Foundation of China (82001673 and 82272860).
Collapse
Affiliation(s)
- Fangyuan Jiang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Lijuan Wang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China; Centre for Global Health, Usher Institute, the University of Edinburgh, Edinburgh, UK
| | - Haochao Ying
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Sun
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhui Zhao
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Lu
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Zilong Bian
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Chen
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Aiping Fang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Xuehong Zhang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Susanna C Larsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Unit of Medical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuai Yuan
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xue Li
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
22
|
Yang MY, Liu YJ, Zheng MH, Pan T, Li ZY, Gong BF, Fan HY. Anti-adipogenesis and anti-obesity potential of alliin mediated by modulating glycolipid metabolism via activating PPARγ signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8707-8723. [PMID: 38829385 DOI: 10.1007/s00210-024-03181-w] [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: 02/19/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Garlic exhibits hypolipidemic, hypoglycemic, and cardiovascular benefits. The inconsistent results of garlic preparations on adipogenesis have caused more confusion in the public and academia. The compounds responsible for the anti-adipogenesis effect of garlic remain unknown. The present study aimed to verify the real anti-adipogenesis and anti-obesity component in garlic and explored its possible effects in metabolic syndrome. We verified the real anti-adipogenesis and anti-obesity components of garlic in 3T3-L1 preadipocytes and a 10-week-high fat diet (HFD)-induced obese mice. In vitro, two water-soluble and four typical lipid-soluble compounds of garlic were tested for their anti-adipogenesis. Then, the water-soluble compound, alliin, and two processing methods produced garlic oils, were evaluated in vivo study. Mice received oral administration of alliin (25 mg/kg) and garlic oils (15 mg/kg) daily for 8 weeks. Serum lipids, parameters of obesity, and indicators involved in regulating glycolipid metabolism were examined. Our findings confirmed that both water-soluble and lipid-soluble organosulfur compounds of garlic contributed to garlic's anti-adipogenesis effect, in which water-soluble sulfides, especially alliin, exhibited greater potency. Alliin possessed potent effects of anti-obesity and improvement in glucose and lipid metabolism in HFD-induced obese mice. Alliin mediated these effects partly attributed to its modulation of enzymatic activities within glycolipid metabolism and activating PPARγ signaling pathway. In contrast to odorous lipid-soluble sulfides, alliin is odorless, stable, and safe, and is an ideal nutraceutical or even medicinal candidates for the treatment of metabolic diseases. Alliin could be used to standardize the quality of garlic products.
Collapse
Affiliation(s)
- Ming-Yan Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong Province, China
| | - Yu-Jing Liu
- Department of Pharmacy, Fushan District People's Hospital of Yantai City, No. 111 Gangchengxidajie, Fushan District, Yantai, 265500, Shandong Province, China
| | - Mei-Hua Zheng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong Province, China
| | - Tao Pan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong Province, China
| | - Zhen-Yuan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong Province, China
| | - Bai-Fang Gong
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong Province, China
| | - Hua-Ying Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong Province, China.
| |
Collapse
|
23
|
Noh SG, Kim HW, Kim S, Chung KW, Jung YS, Yoon JH, Yu BP, Lee J, Chung HY. Senoinflammation as the underlying mechanism of aging and its modulation by calorie restriction. Ageing Res Rev 2024; 101:102503. [PMID: 39284417 DOI: 10.1016/j.arr.2024.102503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Senoinflammation is characterized by an unresolved low-grade inflammatory process that affects multiple organs and systemic functions. This review begins with a brief overview of the fundamental concepts and frameworks of senoinflammation. It is widely involved in the aging of various organs and ultimately leads to progressive systemic degeneration. Senoinflammation underlying age-related inflammation, is causally related to metabolic dysregulation and the formation of senescence-associated secretory phenotype (SASP) during aging and age-related diseases. This review discusses the biochemical evidence and molecular biology data supporting the concept of senoinflammation and its regulatory processes, highlighting the anti-aging and anti-inflammatory effects of calorie restriction (CR). Experimental data from CR studies demonstrated effective suppression of various pro-inflammatory cytokines and chemokines, lipid accumulation, and SASP during aging. In conclusion, senoinflammation represents the basic mechanism that creates a microenvironment conducive to aging and age-related diseases. Furthermore, it serves as a potential therapeutic target for mitigating aging and age-related diseases.
Collapse
Affiliation(s)
- Sang Gyun Noh
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Hyun Woo Kim
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Seungwoo Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Ki Wung Chung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Young-Suk Jung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jeong-Hyun Yoon
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jaewon Lee
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea.
| | - Hae Young Chung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea.
| |
Collapse
|
24
|
Guo Z, Yao Z, Huang B, Wu D, Li Y, Chen X, Lu Y, Wang L, Lv W. MAFLD-related hepatocellular carcinoma: Exploring the potent combination of immunotherapy and molecular targeted therapy. Int Immunopharmacol 2024; 140:112821. [PMID: 39088919 DOI: 10.1016/j.intimp.2024.112821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common cause of cancer-related mortality and morbidity globally, and with the prevalence of metabolic-related diseases, the incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) related hepatocellular carcinoma (MAFLD-HCC) continues to rise with the limited efficacy of conventional treatments, which has created a major challenge for HCC surveillance. Immune checkpoint inhibitors (ICIs) and molecularly targeted drugs offer new hope for advanced MAFLD-HCC, but the evidence for the use of both types of therapy in this type of tumour is still insufficient. Theoretically, the combination of immunotherapy, which awakens the body's anti-tumour immunity, and targeted therapies, which directly block key molecular events driving malignant progression in HCC, is expected to produce synergistic effects. In this review, we will discuss the progress of immunotherapy and molecular targeted therapy in MAFLD-HCC and look forward to the opportunities and challenges of the combination therapy.
Collapse
Affiliation(s)
- Ziwei Guo
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People 's Hospital, Beijing 100044, China
| | - Bohao Huang
- Beijing University of Chinese Medicine, Beijing 100105, China
| | - Dongjie Wu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yanbo Li
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiaohan Chen
- Department of Hematology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanping Lu
- Department of Hepatology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518100, China.
| | - Li Wang
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Wenliang Lv
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
25
|
Weiss U, Mungo E, Haß M, Benning D, Gurke R, Hahnefeld L, Dorochow E, Schlaudraff J, Schmid T, Kuntschar S, Meyer S, Medert R, Freichel M, Geisslinger G, Niederberger E. Knock-Out of IKKepsilon Ameliorates Atherosclerosis and Fatty Liver Disease by Alterations of Lipid Metabolism in the PCSK9 Model in Mice. Int J Mol Sci 2024; 25:10721. [PMID: 39409049 PMCID: PMC11476531 DOI: 10.3390/ijms251910721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
The inhibitor-kappaB kinase epsilon (IKKε) represents a non-canonical IκB kinase that modulates NF-κB activity and interferon I responses. Inhibition of this pathway has been linked with atherosclerosis and metabolic dysfunction-associated steatotic liver disease (MASLD), yet the results are contradictory. In this study, we employed a combined model of hepatic PCSK9D377Y overexpression and a high-fat diet for 16 weeks to induce atherosclerosis and liver steatosis. The development of atherosclerotic plaques, serum lipid concentrations, and lipid metabolism in the liver and adipose tissue were compared between wild-type and IKKε knock-out mice. The formation and progression of plaques were markedly reduced in IKKε knockout mice, accompanied by reduced serum cholesterol levels, fat deposition, and macrophage infiltration within the plaque. Additionally, the development of a fatty liver was diminished in these mice, which may be attributed to decreased levels of multiple lipid species, particularly monounsaturated fatty acids, triglycerides, and ceramides in the serum. The modulation of several proteins within the liver and adipose tissue suggests that de novo lipogenesis and the inflammatory response are suppressed as a consequence of IKKε inhibition. In conclusion, our data suggest that the knockout of IKKε is involved in mechanisms of both atherosclerosis and MASLD. Inhibition of this pathway may therefore represent a novel approach to the treatment of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ulrike Weiss
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Eleonora Mungo
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Michelle Haß
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Denis Benning
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Robert Gurke
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Lisa Hahnefeld
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Erika Dorochow
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Jessica Schlaudraff
- Goethe University Frankfurt, Faculty of Medicine, Institute of Neuroanatomy, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany;
| | - Tobias Schmid
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Silvia Kuntschar
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Sofie Meyer
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Rebekka Medert
- Institute of Pharmacology, Ruprechts-Karl University Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (R.M.); (M.F.)
| | - Marc Freichel
- Institute of Pharmacology, Ruprechts-Karl University Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (R.M.); (M.F.)
| | - Gerd Geisslinger
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Ellen Niederberger
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
26
|
Liang M, Dong S, Guo Y, Zhang Y, Xiao X, Ma J, Jiang X, Yu W. Exploration of the potential mechanism of aqueous extract of Artemisia capillaris for the treatment of non-alcoholic fatty liver disease based on network pharmacology and experimental verification. J Pharm Pharmacol 2024; 76:1328-1339. [PMID: 39186724 DOI: 10.1093/jpp/rgae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 08/06/2024] [Indexed: 08/28/2024]
Abstract
OBJECTIVES Non-alcoholic fatty liver disease (NAFLD) is a nutritional and metabolic disease with a high prevalence today. Artemisia capillaris has anti-inflammatory, antioxidant, and other effects. However, the mechanism of A. capillaris in treating NAFLD is still poorly understood. METHODS This study explored the mechanism of A. capillaris in the treatment of NAFLD through network pharmacology and molecular docking, and verified the results through in vivo experiments using a high-fat diet-induced mouse model and in vitro experiments using an oleic acid-induced HepG2 cell model. KEY FINDINGS Aqueous extract of A. capillaris (AEAC) can reduce blood lipids, reduce liver lipid accumulation and liver inflammation in NAFLD mice, and improve NAFLD. Network pharmacology analysis revealed that 51 drug ingredients in A. capillaris correspond to 370 targets that act on NAFLD. GEO data mining obtained 93 liver differentially expressed genes related to NAFLD. In the UHPLC-MS detection results, 36 components were characterized and molecular docked with JNK. Verified in vitro and in vivo, the results show that JNK and the phosphorylation levels of IL-6, IL-1β, c-Jun, c-Fos, and CCL2 are key targets and pathways. CONCLUSIONS This study confirmed that AEAC reduces lipid accumulation and inflammation in the liver of NAFLD mice by inhibiting the JNK/AP-1 pathway.
Collapse
Affiliation(s)
- Meng Liang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Siyu Dong
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yi Guo
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yuyi Zhang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiao Xiao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jun Ma
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Heilongjiang Provincial Key Laboratory for Prevention and Control of Common Animal Diseases, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Heilongjiang Provincial Key Laboratory for Prevention and Control of Common Animal Diseases, Northeast Agricultural University, Harbin, 150030, China
| |
Collapse
|
27
|
Gu L, Du Y, Liang F. Meta-Analysis and Network Analysis Differentially Detect Various Pro-Inflammatory Mediators and Risk Factors for Type 2 Diabetes in the Elderly. Horm Metab Res 2024; 56:727-736. [PMID: 38195796 DOI: 10.1055/a-2241-5281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Type 2 diabetes (T2D) has a pathophysiological component that includes inflammation. Inflammation-sensitive marker measurement may be helpful in determining the risk of complications for both older T2D patients and the public. This study aimed to investigate the association between blood pro-inflammatory mediators and the characteristics of elderly patients with T2D using meta and network analyses. The Web of Science, Scopus, PubMed, and Cochrane Library databases were selected as study methodology. The Quality in Prognosis Studies (QUIPS) tool in the meta-analysis assessed the studies' methodological quality. The selected studies were statistically analyzed using the META-MAR tool based on the standardized mean difference (SMD). The selected studies included nine examinations involving 6399 old people [+>+55 years old, 65.9+±+4.09 (mean+±+SD)]. The meta-analysis showed that pro-inflammatory mediators (SMD 0.82) and patient-related variables [risk factors (SMD 0.71)] were significantly associated with T2D (p+<+0.05). Subgroup analysis revealed that tumor necrosis factor alpha (TNF-α; SMD 1.08), body mass index (SMD 0.64), high-density lipoprotein (HDL; SMD -0.61), body weight (SMD 0.50), and blood pressure (SMD 1.11) were factors significantly associated with T2D (p+<+0.05). Network analysis revealed that among patient characteristics, diastolic blood pressure and, among inflammatory mediators, leptin were the most closely associated factors with T2D in older adults. Moreover, network analysis showed that TNF-α and systolic blood pressure were most closely associated with leptin. Overall, alternate techniques, such as meta-analysis and network analysis, might identify different markers for T2D in older people. A therapeutic decision-making process needs to consider these differences in advance.
Collapse
Affiliation(s)
- Linlin Gu
- Endocrine Metabolism Department and Geriatric Department, 7th People's Hospital of Chengdu, Chengdu, China
| | - Yue Du
- Endocrine Metabolism Department and Geriatric Department, 7th People's Hospital of Chengdu, Chengdu, China
| | - Fang Liang
- General Medicine Department, 7th People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
28
|
Okuma H, Tsuchiya K. Tissue-specific activation of insulin signaling as a potential target for obesity-related metabolic disorders. Pharmacol Ther 2024; 262:108699. [PMID: 39111411 DOI: 10.1016/j.pharmthera.2024.108699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this "healthy adipose tissue expansion" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue.
Collapse
Affiliation(s)
- Hideyuki Okuma
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan
| | - Kyoichiro Tsuchiya
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan.
| |
Collapse
|
29
|
Huang F, Gao J, Li A, Mizokami A, Matsuda M, Aoki K, Katagiri T, Kawakubo-Yasukochi T, Jimi E. Activation of NF-κB signaling regulates ovariectomy-induced bone loss and weight gain. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167320. [PMID: 38936515 DOI: 10.1016/j.bbadis.2024.167320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
Postmenopausal women experience bone loss and weight gain. To date, crosstalk between estrogen receptor signals and nuclear factor-κB (NF-κB) has been reported, and estrogen depletion enhances bone resorption by osteoclasts via NF-κB activation. However, it is unclear when and in which tissues NF-κB is activated after menopause, and how NF-κB acts as a common signaling molecule for postmenopausal weight gain and bone loss. Therefore, we examined the role of NF-κB in bone and energy metabolism following menopause. NF-κB reporter mice, which can be used to measure NF-κB activation in vivo, were ovariectomized (OVX) and the luminescence intensity after OVX increased in the metaphyses of the long bones and perigonadal white adipose tissue, but not in the other tissues. OVX was performed on wild-type (WT) and p65 mutant knock-in (S534A) mice, whose mutation enhances the transcriptional activity of NF-κB. Weight gain with worsening glucose tolerance was significant in S534A mice after OVX compared with those of WT mice. The bone density of the sham group in WT or S534A mice did not change, whereas in the S534A-OVX group it significantly decreased due to the suppression of bone formation and increase in bone marrow adipocytes. Disulfiram, an anti-alcoholic drug, suppressed OVX-induced activation of NF-κB in the metaphyses of long bones and white adipose tissue (WAT), as well as weight gain and bone loss. Overall, the activation of NF-κB in the metaphyses of long bones and WAT after OVX regulates post-OVX weight gain and bone loss.
Collapse
Affiliation(s)
- Fei Huang
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jing Gao
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Aonan Li
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akiko Mizokami
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Miho Matsuda
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazuhiro Aoki
- Department of Basic Oral Health Engineering, Graduate School, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8549, Japan
| | - Takenobu Katagiri
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, Hidaka-shi, Saitama 350-1241, Japan
| | - Tomoyo Kawakubo-Yasukochi
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Eijiro Jimi
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
30
|
Qin Y, Yao S, Wang G, Xu Z, Zhu G, Li Y, Yang P, Wang J, Zou C, Chen C. The Gastric Connection: Serum Gastric Biomarkers, Metabolic Syndrome and Transition in Metabolic Status. J Inflamm Res 2024; 17:6439-6452. [PMID: 39310898 PMCID: PMC11414752 DOI: 10.2147/jir.s475352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Objective Inflammation plays a crucial role in the development of metabolic syndrome (MetS). However, the roles of pepsinogens (PGs) and gastrin, known biomarkers linked to gastric inflammation, in MetS and the transition of MetS status are unclear. This research aimed to explore the relationship between MetS, the transition of MetS status, and levels of gastric biomarkers. Methods This large-scale cross-sectional study included 19162 participants aged 18-80 years between August 2021 and March 2024. Serum levels of the gastric biomarkers PGI, PGII, and gastrin-17 were analyzed using enzyme-linked immunosorbent assay. In addition, the relationship between transitions of MetS status based on 1032 MetS-negative participants from baseline to the second health exam after 2 years was considered. The association between MetS and the transitions of MetS status and gastric biomarkers was analyzed using logistic regression models. Results The prevalence of MetS in the study population was 31.4%, with higher rates in males (35.2%) than females (24.6%). Gastrin-17 levels were markedly elevated in participants with MetS, a trend observed in both genders. In the logistic regression analysis, after adjusting for confounding factors, gastrin-17 levels were strongly and positively correlated with MetS in the entire cohort and in males but not in females. Male participants with MetS had lower levels of PGI and PGII than those without MetS, whereas the opposite trend was observed in females. Logistic regression analysis indicated that PGI and PGII were not independently associated with MetS. During the follow-up of 2 years, 199 (19.28%) of the 1032 MetS-negative participants transitioned to MetS-positive status. As compared to the stable MetS-negative subjects, transition from MetS-negative to MetS-positive was associated with higher levels of gastrin-17, especially in males, but not in females. Conclusion Gastrin-17 is a promising biomarker for MetS, exhibiting potential utility in monitoring the transition of MetS status and revealing gender difference.
Collapse
Affiliation(s)
- Yuexiang Qin
- Department of Health Management Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
- Hunan Chronic Disease Health Management Medical Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
| | - Shanhu Yao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
- Key Laboratory of Medical Information Research, Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
| | - Gaoyuan Wang
- Department of Nephrology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Ziyuan Xu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Gangcai Zhu
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, People’s Republic of China
| | - Ying Li
- Department of Health Management Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
- Hunan Chronic Disease Health Management Medical Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
| | - Pingting Yang
- Department of Health Management Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
- Hunan Chronic Disease Health Management Medical Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
| | - Jiangang Wang
- Department of Health Management Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
- Hunan Chronic Disease Health Management Medical Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, 410013, People’s Republic of China
| | - Chunbo Zou
- Department of Nephrology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Changhan Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, People’s Republic of China
| |
Collapse
|
31
|
Wensveen FM, Šestan M, Polić B. The immunology of sickness metabolism. Cell Mol Immunol 2024; 21:1051-1065. [PMID: 39107476 PMCID: PMC11364700 DOI: 10.1038/s41423-024-01192-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/29/2024] [Indexed: 09/01/2024] Open
Abstract
Everyone knows that an infection can make you feel sick. Although we perceive infection-induced changes in metabolism as a pathology, they are a part of a carefully regulated process that depends on tissue-specific interactions between the immune system and organs involved in the regulation of systemic homeostasis. Immune-mediated changes in homeostatic parameters lead to altered production and uptake of nutrients in circulation, which modifies the metabolic rate of key organs. This is what we experience as being sick. The purpose of sickness metabolism is to generate a metabolic environment in which the body is optimally able to fight infection while denying vital nutrients for the replication of pathogens. Sickness metabolism depends on tissue-specific immune cells, which mediate responses tailored to the nature and magnitude of the threat. As an infection increases in severity, so do the number and type of immune cells involved and the level to which organs are affected, which dictates the degree to which we feel sick. Interestingly, many alterations associated with metabolic disease appear to overlap with immune-mediated changes observed following infection. Targeting processes involving tissue-specific interactions between activated immune cells and metabolic organs therefore holds great potential for treating both people with severe infection and those with metabolic disease. In this review, we will discuss how the immune system communicates in situ with organs involved in the regulation of homeostasis and how this communication is impacted by infection.
Collapse
Affiliation(s)
| | - Marko Šestan
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
32
|
Leungsuwan DS, Chandran M. Bone Fragility in Diabetes and its Management: A Narrative Review. Drugs 2024; 84:1111-1134. [PMID: 39103693 DOI: 10.1007/s40265-024-02078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Bone fragility is a serious yet under-recognised complication of diabetes mellitus (DM) that is associated with significant morbidity and mortality. Multiple complex pathophysiological mechanisms mediating bone fragility amongst DM patients have been proposed and identified. Fracture risk in both type 1 diabetes (T1D) and type 2 diabetes (T2D) continues to be understated and underestimated by conventional risk assessment tools, posing an additional challenge to the identification of at-risk patients who may benefit from earlier intervention or preventive strategies. Over the years, an increasing body of evidence has demonstrated the efficacy of osteo-pharmacological agents in managing skeletal fragility in DM. This review seeks to elaborate on the risk of bone fragility in DM, the underlying pathogenesis and skeletal alterations, the approach to fracture risk assessment in DM, management strategies and therapeutic options.
Collapse
Affiliation(s)
| | - Manju Chandran
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, 20 College Road, ACADEMIA, Singapore, 169856, Singapore.
- DUKE NUS Medical School, Singapore, Singapore.
| |
Collapse
|
33
|
Chowdhury AS, Palui R, Pramanik S, Mondal S. Glycemic variability in chronic calcific pancreatitis with diabetes mellitus and its possible determinants. Diabetes Metab Syndr 2024; 18:103100. [PMID: 39142092 DOI: 10.1016/j.dsx.2024.103100] [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: 09/05/2023] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
AIMS To study glycemic patterns and variability in patients with pancreatic diabetes or type 3c Diabetes mellitus (DM) due to chronic pancreatitis and its subtypes and assess the role of pancreatic enzyme replacement therapy (ERT) in modulating glycemic variability. METHODS Patients having type 3c DM due to chronic pancreatitis, and on insulin underwent Flash continuous-glucose-monitoring (CGM) for 14 days. Parameters were compared between patients with fibrocalculous pancreatic diabetes (FCPD) and non-FCPD-chronic calcific pancreatitis (non-FCPD) and between the recipients and non-recipients of pancreatic enzyme-replacement-therapy (ERT). RESULTS Out of 54 patients with pancreatic diabetes, 35 patients had chronic calcific pancreatitis. They underwent CGM, median HbA1c 9.20 % (77 mmol/mol) and mean Time-In-Range (TIR) being 41.21 % (23.48). Only 5 (15.2 %) patients achieved target TIR>70 % while 16 (48.5 %) patients had high glycemic-variability [Coefficient-of-variation (CV) > 36 %]. Patients with FCPD (n = 14) had higher hypoglycemia-indices like Time-Below-Range (18.92 % vs 8.20 %; p = 0.03) and Low-Blood-Glucose-Index (18.14 % vs 6.04 %; p = 0.02) compared to non-FCPD (n = 21). HbA1c% and hyperglycemic excursions were similar in both groups. Recipients of ERT (n = 20) had lower glycemic-variability [Standard Deviation (SD) 52.15 % vs 68.14 % and CV 32.59 % vs 41.79 %, p < 0.05 for both) than non-recipients. ERT-recipients had no serious hypoglycemia within the 14 days. On subgroup analysis, lower glycemic-variability and hypoglycemia with ERT were seen only in FCPD but not in non-FCPD subgroup (50.13 vs 77.91, 30.09 vs 48.36 for SD and CV respectively, p < 0.05). CONCLUSION Patients with type 3c DM due to chronic pancreatitis have high frequency of hyperglycemic and hypoglycemic excursions, with those with FCPD having a particularly higher risk of hypoglycemia and glycemic-variability. Those receiving pancreatic ERT had lesser glycemic variability and hypoglycemia. The small sample size and lack of objective markers of documentation of exocrine pancreatic insufficiency like fecal elastase highlight the need for further larger studies in this field.
Collapse
Affiliation(s)
| | - Rajan Palui
- Department of Endocrinology, The Mission Hospital, Durgapur, West Bengal, India.
| | | | - Sunetra Mondal
- Department of Endocrinology and Metabolism, HealthWorld hospitals, Durgapur, India.
| |
Collapse
|
34
|
Huang C, Chen X, Ouyang Z, Meng L, Liu J, Pang Q, Fan R. Bisphenol a accelerates the glucolipotoxicity-induced dysfunction of rat insulinoma cell lines: An implication for a potential risk of environmental bisphenol a exposure for individuals susceptible to type 2 diabetes. Toxicol In Vitro 2024; 99:105866. [PMID: 38844119 DOI: 10.1016/j.tiv.2024.105866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Epidemiological studies have suggested a correlation between bisphenol A (BPA) and type 2 diabetes (T2DM). The effects of BPA on β-cell dysfunction may reveal the risks from an in vitro perspective. We used the rat insulinoma (INS-1) cell lines (a type of β-cells) to set up normal or damaged models (DM), which were exposed to various concentrations of BPA (0.001, 0.01, 0.1, 1, 10 and 100 μM). An increase in reactive oxygen species (ROS) and apoptosis, and a decrease in cell viability were observed in INS-1 cells exposed to high doses of BPA for 48 h. Interestingly, exposure to lower doses of BPA for 24 h resulted in increased ROS levels and apoptosis rates in INS-1 in the DM group, along with decreased cell viability, suggesting that BPA exerts toxicity to INS-1 cells, particularly to the DM group. Insulin levels and Glut2 expression, glucose consumption, intracellular Ca2+ and insulin secretion were increased in INS-1 cells after 48 h exposure to high dose of BPA. Stronger effects were observed in the DM group, even those exposed to low doses of BPA for 24 h. Moreover, BPA inhibited high glucose-stimulated insulin secretion in these cells. Our research suggests that low doses of BPA exacerbate the dysfunction caused by glucolipotoxicity, implying environmental BPA exposure poses a risk for individuals with prediabetes or T2DM.
Collapse
Affiliation(s)
- Chengmeng Huang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaolin Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zedong Ouyang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Lingxue Meng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jian Liu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Qihua Pang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
35
|
Arvanitakis K, Chatzikalil E, Kalopitas G, Patoulias D, Popovic DS, Metallidis S, Kotsa K, Germanidis G, Koufakis T. Metabolic Dysfunction-Associated Steatotic Liver Disease and Polycystic Ovary Syndrome: A Complex Interplay. J Clin Med 2024; 13:4243. [PMID: 39064282 PMCID: PMC11278502 DOI: 10.3390/jcm13144243] [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: 06/08/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and polycystic ovary syndrome (PCOS) are prevalent conditions that have been correlated with infertility through overlapped pathophysiological mechanisms. MASLD is associated with metabolic syndrome and is considered among the major causes of chronic liver disease, while PCOS, which is characterized by ovulatory dysfunction and hyperandrogenism, is one of the leading causes of female infertility. The pathophysiological links between PCOS and MASLD have not yet been fully elucidated, with insulin resistance, hyperandrogenemia, obesity, and dyslipidemia being among the key pathways that contribute to liver lipid accumulation, inflammation, and fibrosis, aggravating liver dysfunction. On the other hand, MASLD exacerbates insulin resistance and metabolic dysregulation in women with PCOS, creating a vicious cycle of disease progression. Understanding the intricate relationship between MASLD and PCOS is crucial to improving clinical management, while collaborative efforts between different medical specialties are essential to optimize fertility and liver health outcomes in individuals with MASLD and PCOS. In this review, we summarize the complex interplay between MASLD and PCOS, highlighting the importance of increasing clinical attention to the prevention, diagnosis, and treatment of both entities.
Collapse
Affiliation(s)
- Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Elena Chatzikalil
- Athens Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Georgios Kalopitas
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Dimitrios Patoulias
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Djordje S. Popovic
- Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Centre of Vojvodina, 21000 Novi Sad, Serbia;
- Medical Faculty, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Symeon Metallidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Division of Endocrinology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece;
| | - Kalliopi Kotsa
- Division of Endocrinology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece;
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Theocharis Koufakis
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| |
Collapse
|
36
|
Harvei S, Skogen V, Egelandsdal B, Birkeland S, Paulsen JE, Carlsen H. Chronic oral LPS administration does not increase inflammation or induce metabolic dysregulation in mice fed a western-style diet. Front Nutr 2024; 11:1376493. [PMID: 39077160 PMCID: PMC11284168 DOI: 10.3389/fnut.2024.1376493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Lipopolysaccharides (LPS) present in the intestine are suggested to enter the bloodstream after consumption of high-fat diets and cause systemic inflammation and metabolic dysregulation through a process named "metabolic endotoxemia." This study aimed to determine the role of orally administered LPS to mice in the early stage of chronic low-grade inflammation induced by diet. Methods We supplemented the drinking water with E. coli derived LPS to mice fed either high-fat Western-style diet (WSD) or standard chow (SC) for 7 weeks (n = 16-17). Body weight was recorded weekly. Systemic inflammatory status was assessed by in vivo imaging of NF-κB activity at different time points, and glucose dysregulation was assessed by insulin sensitivity test and glucose tolerance test near the end of the study. Systemic LPS exposure was estimated indirectly via quantification of LPS-binding protein (LBP) and antibodies against LPS in plasma, and directly using an LPS-sensitive cell reporter assay. Results and discussion Our results demonstrate that weight development and glucose regulation are not affected by LPS. We observed a transient LPS dependent upregulation of NF-κB activity in the liver region in both diet groups, a response that disappeared within the first week of LPS administration and remained low during the rest of the study. However, WSD fed mice had overall a higher NF-κB activity compared to SC fed mice at all time points independent of LPS administration. Our findings indicate that orally administered LPS has limited to no impact on systemic inflammation and metabolic dysregulation in mice fed a high-fat western diet and we question the capability of intestinally derived LPS to initiate systemic inflammation through a healthy and uncompromised intestine, even when exposed to a high-fat diet.
Collapse
Affiliation(s)
- Silje Harvei
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Vemund Skogen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Bjørg Egelandsdal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Signe Birkeland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Jan Erik Paulsen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, As, Norway
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| |
Collapse
|
37
|
Wang L, Li Y, Li R, Luan J, Cao K, Liu T, Hu H, Chen S, Bu L, Liu L, Wang H, Lu Q. Diverse associations between pancreatic intra-, inter-lobular fat and the development of type 2 diabetes in overweight or obese patients. Front Nutr 2024; 11:1421032. [PMID: 39021593 PMCID: PMC11252058 DOI: 10.3389/fnut.2024.1421032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Pancreatic fat is associated with obesity and type 2 diabetes mellitus (T2DM); however, the relationship between different types of pancreatic fat and diabetes status remains unclear. Therefore, we aimed to determine the potential of different types of pancreatic fat accumulation as a risk factor for T2DM in overweight or obese patients. In total, 104 overweight or obese patients were recruited from January 2020 to December 2022. The patients were divided into three groups: normal glucose tolerance (NGT), impaired fasting glucose or glucose tolerance (IFG/IGT), and T2DM. mDixon magnetic resonance imaging (MRI) was used to detect pancreatic fat in all three groups of patients. The pancreatic head fat (PHF), body fat (PBF), and tail fat (PTF) in the IFG/IGT group were 21, 20, and 31% more than those in the NGT group, respectively. PHF, PBF, and PTF were positively associated with glucose metabolic dysfunction markers in the NGT group, and inter-lobular fat volume (IFV) was positively associated with these markers in the IFG/IGT group. The areas under the receiver operating characteristic curves for PHF, PBF, and PTF (used to evaluate their diagnostic potential for glucose metabolic dysfunction) were 0.73, 0.73, and 0.78, respectively, while those for total pancreatic volume (TPV), pancreatic parenchymal volume, IFV, and IFV/TPV were 0.67, 0.67, 0.66, and 0.66, respectively. These results indicate that intra-lobular pancreatic fat, including PHF, PTF, and PBF, may be a potential independent risk factor for the development of T2DM. Additionally, IFV exacerbates glucose metabolic dysfunction. Intra-lobular pancreatic fat indices were better than IFV for the diagnosis of glucose metabolic dysfunction.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yinghao Li
- Physics Department & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Renfeng Li
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Jinwen Luan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Kaiming Cao
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Tiancheng Liu
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Haiyang Hu
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Shanshan Chen
- College of Medical Imaging, Shanghai University of Medicine and Health Science, Shanghai, China
| | - Le Bu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Medicine School of Tongji University, Shanghai, China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hongzhi Wang
- Physics Department & Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Qing Lu
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
38
|
Elahi R, Nazari M, Mohammadi V, Esmaeilzadeh K, Esmaeilzadeh A. IL-17 in type II diabetes mellitus (T2DM) immunopathogenesis and complications; molecular approaches. Mol Immunol 2024; 171:66-76. [PMID: 38795686 DOI: 10.1016/j.molimm.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 05/28/2024]
Abstract
Chronic inflammation has long been considered the characteristic feature of type II diabetes mellitus (T2DM) Immunopathogenesis. Pro-inflammatory cytokines are considered the central drivers of the inflammatory cascade leading to β-cell dysfunction and insulin resistance (IR), two major pathologic events contributing to T2DM. Analyzing the cytokine profile of T2DM patients has also introduced interleukin-17 (IL-17) as an upstream regulator of inflammation, regarding its role in inducing the nuclear factor-kappa B (NF-κB) pathway. In diabetic tissues, IL-17 induces the expression of inflammatory cytokines and chemokines. Hence, IL-17 can deteriorate insulin signaling and β-cell function by activating the JNK pathway and inducing infiltration of neutrophils into pancreatic islets, respectively. Additionally, higher levels of IL-17 expression in patients with diabetic complications compared to non-complicated individuals have also proposed a role for IL-17 in T2DM complications. Here, we highlight the role of IL-17 in the Immunopathogenesis of T2DM and corresponding pathways, recent advances in preclinical and clinical studies targeting IL-17 in T2DM, and corresponding challenges and possible solutions.
Collapse
Affiliation(s)
- Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Vahid Mohammadi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kimia Esmaeilzadeh
- Department of Medical Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
39
|
Liu C, Guo X, Wang B, Meng T, Li C, Zhou Y, Fu J. Association between Cardiometabolic Index and Cognitive Function: A Cross-Sectional Study in a Diabetic-Based Population. Dement Geriatr Cogn Disord 2024; 53:237-247. [PMID: 38843782 DOI: 10.1159/000539482] [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: 04/08/2024] [Accepted: 05/20/2024] [Indexed: 07/12/2024] Open
Abstract
INTRODUCTION Diabetes is a significant risk factor for cognitive impairment. Therefore, early identification of cognitive impairment in diabetic patients is particularly important. The aim of this study was to assess the relationship between Cardiometabolic index (CMI) and cognitive function in a diabetic population. METHODS A cross-sectional study was conducted by collecting information from the National Health and Nutrition Examination Survey (NHANES) 2011-2014. Multiple linear regression models were used to investigate the correlation between CMI and low cognitive function in a diabetic population. Threshold effects analysis and fitted smoothing curves were used to describe the nonlinear links. Interaction tests and subgroup analyses were also performed. RESULTS A total of 1,050 people participated in this study, including 561 men and 489 women. In the fully corrected model, CMI was positively associated with low cognitive performance as assessed by CERAD Word List Learning Test (CERAD W-L), Animal Fluency Test (AFT), and Digit Symbol Substitution Test (DSST) (OR = 1.37 [1.14, 1.72], p = 7.4 × 10-3), (OR = 1.21 [1.04, 1.51], p = 1.26 × 10-2), and (OR = 1.27 [1.08, 1.63], p = 2.53 × 10-2). Our study found that diabetic patients with higher CMI were at greater risk of developing low cognitive function. The effect of the subgroups on the positive association of CMI with cognitive impairment was not significant. A non-linear association between low cognitive performance and CMI was determined by CERAD W-L, AFT, and DSST (log-likelihood ratio <5 × 10-2). In addition, our also study found that CMI was a better predictor of cognitive impairment in diabetes than weight-adjusted waist index (WWI). CONCLUSION Increased CMI is associated with an increased risk of cognitive impairment in people with diabetes. CMI can be used as a new anthropometric measure for predicting cognitive impairment in diabetes, with stronger predictive power than WWI.
Collapse
Affiliation(s)
- Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China,
| | - Xinyi Guo
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- The First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiamei Fu
- The First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
40
|
Hasanian-Langroudi F, Ghasemi A, Hedayati M, Siadat SD, Tohidi M. Novel Insight into the Effect of Probiotics in the Regulation of the Most Important Pathways Involved in the Pathogenesis of Type 2 Diabetes Mellitus. Probiotics Antimicrob Proteins 2024; 16:829-844. [PMID: 37162668 DOI: 10.1007/s12602-023-10056-8] [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: 03/02/2023] [Indexed: 05/11/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is considered one of the most common disorders worldwide. Although several treatment modalities have been developed, the existing interventions have not yielded the desired results. Therefore, researchers have focused on finding treatment choices with low toxicity and few adverse effects that could control T2DM efficiently. Various types of research on the role of gut microbiota in developing T2DM and its related complications have led to the growing interest in probiotic supplementation. Several properties make these organisms unique in terms of human health, including their low cost, high reliability, and good safety profile. Emerging evidence has demonstrated that three of the most important signaling pathways, including nuclear factor kappa B (NF-κB), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), and nuclear factor erythroid 2-related factor 2 (Nrf2), which involved in the pathogenesis of T2DM, play key functions in the effects of probiotics on this disease. Hence, we will focus on the clinical applications of probiotics in the management of T2DM. Then, we will also discuss the roles of the involvement of various probiotics in the regulation of the most important signaling pathways (NF-κB, PI3K/Akt, and Nrf2) involved in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Farzaneh Hasanian-Langroudi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box, Tehran, 19395-4763, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Maryam Tohidi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box, Tehran, 19395-4763, Iran.
| |
Collapse
|
41
|
Gotoh K, Satoh F, Uesugi H, Noguchi T, Matsuda N, Sada K, Miyamoto S, Ozeki Y, Yoshida Y, Okamoto M, Masaki T, Kawabe T, Shibata H. Susceptibility to recombinant SARS-CoV-2 spike protein entry in the lungs of high-fat diet-induced obese mice. FASEB J 2024; 38:e23656. [PMID: 38752523 DOI: 10.1096/fj.202301864rr] [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: 09/21/2023] [Revised: 03/26/2024] [Accepted: 04/24/2024] [Indexed: 07/16/2024]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Obesity is a major risk factor for the development of COVID-19. Angiotensin-converting enzyme 2 (ACE2) is an essential receptor for cell entry of SARS-CoV-2. The receptor-binding domain of the S1 subunit (S1-RBD protein) in the SARS-CoV-2 spike glycoprotein binds to ACE2 on host cells, through which the virus enters several organs, including the lungs. Considering these findings, recombinant ACE2 might be utilized as a decoy protein to attenuate SARS-CoV-2 infection. Here, we examined whether obesity increases ACE2 expression in the lungs and whether recombinant ACE2 administration diminishes the entry of S1-RBD protein into lung cells. We observed that high-fat diet-induced obesity promoted ACE2 expression in the lungs by increasing serum levels of LPS derived from the intestine. S1-RBD protein entered the lungs specifically through ACE2 expressed in host lungs and that the administration of recombinant ACE2 attenuated this entry. We conclude that obesity makes hosts susceptible to recombinant SARS-CoV-2 spike proteins due to elevated ACE2 expression in lungs, and this model of administering S1-RBD protein can be applied to new COVID-19 treatments.
Collapse
Affiliation(s)
- Koro Gotoh
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Fumiko Satoh
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Honami Uesugi
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Takaaki Noguchi
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Naoki Matsuda
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kentaro Sada
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Shotaro Miyamoto
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yoshinori Ozeki
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yuichi Yoshida
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Mitsuhiro Okamoto
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Takayuki Masaki
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | | | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
42
|
Yan S, Santoro A, Niphakis MJ, Pinto AM, Jacobs CL, Ahmad R, Suciu RM, Fonslow BR, Herbst-Graham RA, Ngo N, Henry CL, Herbst DM, Saghatelian A, Kahn BB, Rosen ED. Inflammation causes insulin resistance in mice via interferon regulatory factor 3 (IRF3)-mediated reduction in FAHFA levels. Nat Commun 2024; 15:4605. [PMID: 38816388 PMCID: PMC11139994 DOI: 10.1038/s41467-024-48220-5] [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: 11/08/2022] [Accepted: 04/24/2024] [Indexed: 06/01/2024] Open
Abstract
Obesity-induced inflammation causes metabolic dysfunction, but the mechanisms remain elusive. Here we show that the innate immune transcription factor interferon regulatory factor (IRF3) adversely affects glucose homeostasis through induction of the endogenous FAHFA hydrolase androgen induced gene 1 (AIG1) in adipocytes. Adipocyte-specific knockout of IRF3 protects male mice against high-fat diet-induced insulin resistance, whereas overexpression of IRF3 or AIG1 in adipocytes promotes insulin resistance on a high-fat diet. Furthermore, pharmacological inhibition of AIG1 reversed obesity-induced insulin resistance and restored glucose homeostasis in the setting of adipocyte IRF3 overexpression. We, therefore, identify the adipocyte IRF3/AIG1 axis as a crucial link between obesity-induced inflammation and insulin resistance and suggest an approach for limiting the metabolic dysfunction accompanying obesity.
Collapse
Affiliation(s)
- Shuai Yan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
| | - Anna Santoro
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
| | - Micah J Niphakis
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Antonio M Pinto
- The Salk Institute for Biological Studies, 10010 N. Torey Pines Rd, La Jolla, CA, 92037-1002, USA
| | - Christopher L Jacobs
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Jasim Mohamad Al Bahar St., Kuwait City, Kuwait
| | - Radu M Suciu
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Bryan R Fonslow
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Rachel A Herbst-Graham
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Nhi Ngo
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Cassandra L Henry
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Dylan M Herbst
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Alan Saghatelian
- The Salk Institute for Biological Studies, 10010 N. Torey Pines Rd, La Jolla, CA, 92037-1002, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
- Broad Institute of Harvard and MIT, 320 Charles St., Cambridge, MA, 02141, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA.
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA.
- Broad Institute of Harvard and MIT, 320 Charles St., Cambridge, MA, 02141, USA.
| |
Collapse
|
43
|
Zhang X, Wang Q, Wang Y, Ma C, Zhao Q, Yin H, Li L, Wang D, Huang Y, Zhao Y, Shi X, Li X, Huang C. Interleukin-6 promotes visceral adipose tissue accumulation during aging via inhibiting fat lipolysis. Int Immunopharmacol 2024; 132:111906. [PMID: 38593501 DOI: 10.1016/j.intimp.2024.111906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Age-related visceral obesity could contribute to the development of cardiometabolic complications. The pathogenesis of visceral fat mass accumulation during the aging process remains complex and largely unknown. Interleukin-6 (IL-6) has emerged as one of the prominent inflammaging markers which are elevated in circulation during aging. However, the precise role of IL-6 in regulating age-related visceral adipose tissue accumulation remains uncertain. RESULTS A cross-sectional study including 77 older adults (≥65 years of age) was initially conducted. There was a significant positive association between serum IL-6 levels and visceral fat mass. We subsequently validated a modest but significant elevation in serum IL-6 levels in aged mice. Furthermore, we demonstrated that compared to wildtype control, IL-6 deficiency (IL-6 KO) significantly attenuated the accumulation of visceral adipose tissue during aging. Further metabolic characterization suggested that IL-6 deficiency resulted in improved lipid metabolism parameters and energy expenditure in aged mice. Moreover, histological examinations of adipose depots revealed that the absence of IL-6 ameliorated adipocyte hypertrophy in visceral adipose tissue of aged mice. Mechanically, the ablation of IL-6 could promote the PKA-mediated lipolysis and consequently mitigate lipid accumulation in adipose tissue in aged mice. CONCLUSION Our findings identify a detrimental role of IL-6 during the aging process by promoting visceral adipose tissue accumulation through inhibition of lipolysis. Therefore, strategies aimed at preventing or reducing IL-6 levels may potentially ameliorate age-related obesity and improve metabolism during aging.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Qingxuan Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Yaru Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Chen Ma
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Qing Zhao
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Hongyan Yin
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Long Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Dongmei Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China; Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China
| | - Yinxiang Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Yan Zhao
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Xiulin Shi
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Xuejun Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China.
| | - Caoxin Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China.
| |
Collapse
|
44
|
Hu YA, Guo YX, Huang QF. Rim 18F-fluorodeoxyglucose uptake of hepatic cavernous hemangioma on positron emission tomography/computed tomography: A case report. World J Clin Cases 2024; 12:2243-2247. [PMID: 38808338 PMCID: PMC11129124 DOI: 10.12998/wjcc.v12.i13.2243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/21/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Peripheral FDG accumulation in a hepatic hemangioma presenting in a patient with prolonged fever is rare. Therefore, clinicians should pay close attention to patients with hepatic mass. CASE SUMMARY A 54-year-old woman with a 4-wk history of daily fevers was admitted to our hospital. A whole body 18F-Fluordesoxyglucose (PET-FDG) positron emission tomography/computed tomography (PET/CT) was performed to elucidate the source of the fever. However, whole body 18F-FDG PET/CT raised the suspicion of a malignant lesion because of peripheral FDG accumulation (SUVmax 3.5 g/mL) higher than that of the normal liver parenchyma (SUVmax 1.6 g/mL) surrounding a hypoactive area, and no other abnormalities were showed. Subsequently, the patient underwent liver mass resection. Histopathology showed a hepatic cavernous hemangioma with fatty infiltration around the lesion. The fever disappeared four days after surgery and the patient did not present any complications during follow-up. CONCLUSION Fatty infiltration in the peripheral parts of hepatic cavernous hemangioma may lead to subacute inflammation which further activate the Kupffer cells. This may cause prolonged fever and peripheral rim FDG accumulation on PET/CT.
Collapse
Affiliation(s)
- Yong-An Hu
- Department of Nuclear Medicine, Jinhua Hospital of Zhejiang University, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang Province, China
| | - Ya-Xin Guo
- Department of Nuclear Medicine, Jinhua Hospital of Zhejiang University, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang Province, China
| | - Qi-Feng Huang
- Department of Nuclear Medicine, Jinhua Hospital of Zhejiang University, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang Province, China
| |
Collapse
|
45
|
Luo H, Yang L, Zhang G, Bao X, Ma D, Li B, Cao L, Cao S, Liu S, Bao L, E J, Zheng Y. Whole transcriptome mapping reveals the lncRNA regulatory network of TFP5 treatment in diabetic nephropathy. Genes Genomics 2024; 46:621-635. [PMID: 38536617 DOI: 10.1007/s13258-024-01504-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 02/04/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND TFP5 is a Cdk5 inhibitor peptide, which could restore insulin production. However, the role of TFP5 in diabetic nephropathy (DN) is still unclear. OBJECTIVE This study aims to characterize the transcriptome profiles of mRNA and lncRNA in TFP5-treated DN mice to mine key lncRNAs associated with TFP5 efficacy. METHODS We evaluated the role of TFP5 in DN pathology and performed RNA sequencing in C57BL/6J control mice, C57BL/6J db/db model mice, and TFP5 treatment C57BL/6J db/db model mice. The differentially expressed lncRNAs (DElncRNAs) and mRNAs (DEmRNAs) were analyzed. WGCNA was used to screen hub-gene of TFP5 in treatment of DN. RESULTS Our results showed that TFP5 therapy ameliorated renal tubular injury in DN mice. In addition, compared with the control group, the expression profile of lncRNAs in the model group was significantly disordered, while TFP5 alleviated the abnormal expression of lncRNAs. A total of 67 DElncRNAs shared among the three groups, 39 DElncRNAs showed a trend of increasing in the DN group and decreasing after TFP treatment, while the remaining 28 showed the opposite trend. DElncRNAs were enriched in glycosphingolipid biosynthesis signaling pathways, NF-κB signaling pathways, and complement activation signaling pathways. There were 1028 up-regulated and 1117 down-regulated DEmRNAs in the model group compared to control group, and 123 up-regulated and 153 down-regulated DEmRNAs in the TFP5 group compared to the model group. The DEmRNAs were involved in PPAR and MAPK signaling pathway. We confirmed that MSTRG.28304.1 is a key DElncRNA for TFP5 treatment of DN. TFP5 ameliorated DN maybe by inhibiting MSTRG.28304.1 through regulating the insulin resistance and PPAR signaling pathway. The qRT-PCR results confirmed the reliability of the sequencing data through verifying the expression of ENSMUST00000211209, MSTRG.31814.5, MSTRG.28304.1, and MSTRG.45642.14. CONCLUSION Overall, the present study provides novel insights into molecular mechanisms of TFP5 treatment in DN.
Collapse
Affiliation(s)
- Hongyan Luo
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- The Third Clinical Medical College, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Lirong Yang
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
| | - Guoqing Zhang
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
| | - Xi Bao
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- The Third Clinical Medical College, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Danna Ma
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Bo Li
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- Department of Nephrology Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Li Cao
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
| | - Shilu Cao
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- The Third Clinical Medical College, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Shunyao Liu
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- The Third Clinical Medical College, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Li Bao
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- The Third Clinical Medical College, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Jing E
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yali Zheng
- Department of Nephrology, Ningxia Medical University Affiliated People's Hospital of Autonomous Region, No. 301 Zhengyuan North Street, Yinchuan, 750001, People's Republic of China.
- The Third Clinical Medical College, Ningxia Medical University, Yinchuan, People's Republic of China.
| |
Collapse
|
46
|
Boccatonda A, Del Cane L, Marola L, D’Ardes D, Lessiani G, di Gregorio N, Ferri C, Cipollone F, Serra C, Santilli F, Piscaglia F. Platelet, Antiplatelet Therapy and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Narrative Review. Life (Basel) 2024; 14:473. [PMID: 38672744 PMCID: PMC11051088 DOI: 10.3390/life14040473] [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: 02/28/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is not only related to traditional cardiovascular risk factors like type 2 diabetes mellitus and obesity, but it is also an independent risk factor for the development of cardiovascular disease. MASLD has been shown to be independently related to endothelial dysfunction and atherosclerosis. MASLD is characterized by a chronic proinflammatory response that, in turn, may induce a prothrombotic state. Several mechanisms such as endothelial and platelet dysfunction, changes in the coagulative factors, lower fibrinolytic activity can contribute to induce the prothrombotic state. Platelets are players and addresses of metabolic dysregulation; obesity and insulin resistance are related to platelet hyperactivation. Furthermore, platelets can exert a direct effect on liver cells, particularly through the release of mediators from granules. Growing data in literature support the use of antiplatelet agent as a treatment for MASLD. The use of antiplatelets drugs seems to exert beneficial effects on hepatocellular carcinoma prevention in patients with MASLD, since platelets contribute to fibrosis progression and cancer development. This review aims to summarize the main data on the role of platelets in the pathogenesis of MASLD and its main complications such as cardiovascular events and the development of liver fibrosis. Furthermore, we will examine the role of antiplatelet therapy not only in the prevention and treatment of cardiovascular events but also as a possible anti-fibrotic and anti-tumor agent.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Internal Medicine, Bentivoglio Hospital, AUSL Bologna, 40010 Bentivoglio, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Lorenza Del Cane
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Lara Marola
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Damiano D’Ardes
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, 66100 Chieti, Italy (F.C.)
| | | | - Nicoletta di Gregorio
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Claudio Ferri
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Francesco Cipollone
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, 66100 Chieti, Italy (F.C.)
| | - Carla Serra
- Interventional, Diagnostic and Therapeutic Ultrasound Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Francesca Santilli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, 66100 Chieti, Italy;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
47
|
Wu Y, Ma Y. CCL2-CCR2 signaling axis in obesity and metabolic diseases. J Cell Physiol 2024; 239:e31192. [PMID: 38284280 DOI: 10.1002/jcp.31192] [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: 10/14/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024]
Abstract
Obesity and metabolic diseases, such as insulin resistance, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular ailments, represent formidable global health challenges, bearing considerable implications for both morbidity and mortality rates. It has become increasingly evident that chronic, low-grade inflammation plays a pivotal role in the genesis and advancement of these conditions. The involvement of C-C chemokine ligand 2 (CCL2) and its corresponding receptor, C-C chemokine receptor 2 (CCR2), has been extensively documented in numerous inflammatory maladies. Recent evidence indicates that the CCL2/CCR2 pathway extends beyond immune cell recruitment and inflammation, exerting a notable influence on the genesis and progression of metabolic syndrome. The present review seeks to furnish a comprehensive exposition of the CCL2-CCR2 signaling axis within the context of obesity and metabolic disorders, elucidating its molecular mechanisms, functional roles, and therapeutic implications.
Collapse
Affiliation(s)
- Yue Wu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yanchun Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
48
|
Targher G, Byrne CD, Tilg H. MASLD: a systemic metabolic disorder with cardiovascular and malignant complications. Gut 2024; 73:691-702. [PMID: 38228377 DOI: 10.1136/gutjnl-2023-330595] [Citation(s) in RCA: 80] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/24/2023] [Indexed: 01/18/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has rapidly become the most common chronic liver disease globally and is currently estimated to affect up to 38% of the global adult population. NAFLD is a multisystem disease where systemic insulin resistance and related metabolic dysfunction play a pathogenic role in the development of NAFLD and its most relevant liver-related morbidities (cirrhosis, liver failure and hepatocellular carcinoma) and extrahepatic complications, such as cardiovascular disease (CVD), type 2 diabetes mellitus, chronic kidney disease, and certain types of extrahepatic cancers. In 2023, three large multinational liver associations proposed that metabolic dysfunction-associated steatotic liver disease (MASLD) should replace the term NAFLD; the name chosen to replace non-alcoholic steatohepatitis was metabolic dysfunction-associated steatohepatitis (MASH). Emerging epidemiological evidence suggests an excellent concordance rate between NAFLD and MASLD definitions-that is, ~99% of individuals with NAFLD meet MASLD criteria. In this narrative review, we provide an overview of the literature on (a) the recent epidemiological data on MASLD and the risk of developing CVD and malignant complications, (b) the underlying mechanisms by which MASLD (and factors strongly linked with MASLD) may increase the risk of these extrahepatic complications and (c) the diagnosis and assessment of CVD risk and potential treatments to reduce CVD risk in people with MASLD or MASH.
Collapse
Affiliation(s)
- Giovanni Targher
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore Don Calabria Hospital and Department of Medicine University of Verona, Negrar di Valpolicella (VR), Italy
| | | | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Austria
| |
Collapse
|
49
|
Mei X, Li Y, Zhang X, Zhai X, Yang Y, Li Z, Li L. Maternal Phlorizin Intake Protects Offspring from Maternal Obesity-Induced Metabolic Disorders in Mice via Targeting Gut Microbiota to Activate the SCFA-GPR43 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4703-4725. [PMID: 38349207 DOI: 10.1021/acs.jafc.3c06370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Maternal obesity increases the risk of obesity and metabolic disorders (MDs) in offspring, which can be mediated by the gut microbiota. Phlorizin (PHZ) can improve gut dysbiosis and positively affect host health; however, its transgenerational metabolic benefits remain largely unclear. This study aimed to investigate the potential of maternal PHZ intake in attenuating the adverse impacts of a maternal high-fat diet on obesity-related MDs in dams and offspring. The results showed that maternal PHZ reduced HFD-induced body weight gain and fat accumulation and improved glucose intolerance and abnormal lipid profiles in both dams and offspring. PHZ improved gut dysbiosis by promoting expansion of SCFA-producing bacteria, Akkermansia and Blautia, while inhibiting LPS-producing and pro-inflammatory bacteria, resulting in significantly increased fecal SCFAs, especially butyric acid, and reduced serum lipopolysaccharide levels and intestinal inflammation. PHZ also promoted intestinal GLP-1/2 secretion and intestinal development and enhanced gut barrier function by activating G protein-coupled receptor 43 (GPR43) in the offspring. Antibiotic-treated mice receiving FMT from PHZ-regulated offspring could attenuate MDs induced by receiving FMT from HFD offspring through the gut microbiota to activate the GPR43 pathway. It can be regarded as a promising functional food ingredient for preventing intergenerational transmission of MDs and breaking the obesity cycle.
Collapse
Affiliation(s)
- Xueran Mei
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Yi Li
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney 2052, Australia
| | - Xiaoyu Zhang
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Xiwen Zhai
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney 2052, Australia
| | - Yi Yang
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
| | - Zhengjuan Li
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Liping Li
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| |
Collapse
|
50
|
Chen Y, Chen H, Wang Y, Liu F, Fan X, Shi C, Su X, Tan M, Yang Y, Lin B, Lei K, Qu L, Yang J, Zhu Z, Yuan Z, Xie S, Sun Q, Neculai D, Liu W, Yan Q, Wang X, Shao J, Liu J, Lin A. LncRNA LINK-A Remodels Tissue Inflammatory Microenvironments to Promote Obesity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303341. [PMID: 38145352 PMCID: PMC10933663 DOI: 10.1002/advs.202303341] [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: 05/23/2023] [Revised: 11/09/2023] [Indexed: 12/26/2023]
Abstract
High-fat diet (HFD)-induced obesity is a crucial risk factor for metabolic syndrome, mainly due to adipose tissue dysfunctions associated with it. However, the underlying mechanism remains unclear. This study has used genetic screening to identify an obesity-associated human lncRNA LINK-A as a critical molecule bridging the metabolic microenvironment and energy expenditure in vivo by establishing the HFD-induced obesity knock-in (KI) mouse model. Mechanistically, HFD LINK-A KI mice induce the infiltration of inflammatory factors, including IL-1β and CXCL16, through the LINK-A/HB-EGF/HIF1α feedback loop axis in a self-amplified manner, thereby promoting the adipose tissue microenvironment remodeling and adaptive thermogenesis disorder, ultimately leading to obesity and insulin resistance. Notably, LINK-A expression is positively correlated with inflammatory factor expression in individuals who are overweight. Of note, targeting LINK-A via nucleic acid drug antisense oligonucleotides (ASO) attenuate HFD-induced obesity and metabolic syndrome, pointing out LINK-A as a valuable and effective therapeutic target for treating HFD-induced obesity. Briefly, the results reveale the roles of lncRNAs (such as LINK-A) in remodeling tissue inflammatory microenvironments to promote HFD-induced obesity.
Collapse
Affiliation(s)
- Yu Chen
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Hui Chen
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Ying Wang
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Fangzhou Liu
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Xiao Fan
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Chengyu Shi
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Xinwan Su
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Manman Tan
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Yebin Yang
- The Fourth School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiang310053China
| | - Bangxing Lin
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Kai Lei
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Lei Qu
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Jiecheng Yang
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Zengzhuang Yuan
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)University School of MedicineInternational CampusZhejiang UniversityHainingZhejiang314400China
| | - Shanshan Xie
- The Children's HospitalNational Clinical Research Center for Child HealthZhejiang University School of MedicineHangzhouZhejiang310003China
- Department of Cell BiologyZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Qinming Sun
- Department of BiochemistryDepartment of Cardiology of Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang313000China
- International School of MedicineInternational Institutes of MedicineThe 4th Affiliated Hospital of Zhejiang University School of MedicineYiwuZhejiang322000China
| | - Dante Neculai
- International School of MedicineInternational Institutes of MedicineThe 4th Affiliated Hospital of Zhejiang University School of MedicineYiwuZhejiang322000China
- Department of Cell BiologyDepartment of General Surgery of Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang310016China
| | - Wei Liu
- Department of BiochemistryDepartment of Cardiology of Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang313000China
- International School of MedicineInternational Institutes of MedicineThe 4th Affiliated Hospital of Zhejiang University School of MedicineYiwuZhejiang322000China
| | - Qingfeng Yan
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Xiang Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
- Department of Central LaboratoryThe First People's Hospital of HuzhouHuzhouZhejiang313000China
| | - Jianzhong Shao
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Jian Liu
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)University School of MedicineInternational CampusZhejiang UniversityHainingZhejiang314400China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- Hangzhou Cancer InstitutionAffiliated Hangzhou Cancer HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiang310002China
- College of Medicine and Veterinary MedicineThe University of EdinburghEdinburghEH16 4SBUK
| | - Aifu Lin
- MOE Laboratory of Biosystem Homeostasis and ProtectionCollege of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- International School of MedicineInternational Institutes of MedicineThe 4th Affiliated Hospital of Zhejiang University School of MedicineYiwuZhejiang322000China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058China
- Key Laboratory for Cell and Gene Engineering of Zhejiang ProvinceHangzhouZhejiang310058China
- Future Health LaboratoryInnovation Center of Yangtze River DeltaZhejiang UniversityJiaxingZhejiang314100China
- Key Laboratory of Cancer Prevention and InterventionChina National Ministry of EducationHangzhouZhejiang310009China
| |
Collapse
|