1
|
Dennis KM, Gopal K, Montes Aparicio CN, Zhang JA, Castro-Guarda M, Nicol T, Devereux RM, Carter RD, Azizi SA, Lan T, Purnama U, Carr CA, Simsek G, Gill EK, Swietach P, Sorop O, Heinonen IH, Schianchi F, Luiken JJ, Aksentijevic D, Duncker DJ, Dickinson BC, De Val S, Ussher JR, Fuller W, Heather LC. FoxO1-zDHHC4-CD36 S-Acylation Axis Drives Metabolic Dysfunction in Diabetes. Circ Res 2025; 136:1545-1560. [PMID: 40357580 PMCID: PMC12136392 DOI: 10.1161/circresaha.124.325918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND The fatty acid (FA) transporter CD36 (FA translocase/cluster of differentiation 36) is the gatekeeper of cardiac FA metabolism. Preferential localisation of CD36 to the sarcolemma is one of the initiating cellular responses in the development of muscle insulin resistance and in the type 2 diabetic heart. Post-translational S-acylation controls protein trafficking, and in this study we hypothesised that increased CD36 S-acylation may underpin the preferential sarcolemmal localisation of CD36, driving metabolic and contractile dysfunction in diabetes. METHODS Type 2 diabetes was induced in the rat using high fat diet and a low dose of streptozotocin. Forkhead box O1 (FoxO1) transcriptional regulation of zDHHC4 (zinc finger DHHC-type palmitoyltransferase 4) and subsequent S-acylation of CD36 was assessed using chromatin immunoprecipitation (ChIP) sequencing, ChIP-quantitative polymerase chain reaction, luciferase assays, siRNA (small interfering RNA) and shRNA silencing. RESULTS Type 2 diabetes increased cardiac CD36 S-acylation, CD36 sarcolemmal localisation, FA oxidation rates and triglyceride storage in the diabetic heart. CD36 S-acylation was increased in diabetic rats, db/db mice, diabetic pigs and insulin-resistant human iPSC-derived cardiomyocytes, demonstrating conservation between species. The enzyme responsible for S-acylating CD36, zDHHC4, was transcriptionally upregulated in the diabetic heart, and genetic silencing of zDHHC4 decreased CD36 S-acylation. We identified that zDHHC4 expression is under the regulation of the transcription factor FoxO1. Diabetic mice with cardiomyocyte-specific FoxO1 deletion had decreased cardiac zDHHC4 expression and decreased CD36 S-acylation, which was further confirmed using diabetic mice treated with the FoxO1 inhibitor AS1842856. Pharmacological inhibition of zDHHC enzymes in diabetic hearts decreased CD36 S-acylation, sarcolemmal CD36 content, FA oxidation rates and triglyceride storage, culminating in improved cardiac function in diabetes. Conversely, inhibiting the de-acylating enzymes in control hearts increased CD36 S-acylation, sarcolemmal CD36 content and FA metabolic rates in control hearts, recapitulating the metabolic phenotype seen in diabetic hearts. CONCLUSIONS Activation of the FoxO1-zDHHC4-CD36 S-acylation axis drives metabolic and contractile dysfunction in the type 2 diabetic heart.
Collapse
Affiliation(s)
- Kaitlyn M.J.H. Dennis
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada (K.G., J.R.U.)
| | - Claudia N. Montes Aparicio
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Jiashuo Aaron Zhang
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Marcos Castro-Guarda
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Thomas Nicol
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Ríona M. Devereux
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
- Department of Chemistry (R.M.D.), University of Oxford, United Kingdom
| | - Ryan D. Carter
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Saara-Anne Azizi
- Department of Chemistry, University of Chicago, IL (S.-A.A., T.L., B.C.D.)
| | - Tong Lan
- Department of Chemistry, University of Chicago, IL (S.-A.A., T.L., B.C.D.)
| | - Ujang Purnama
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Carolyn A. Carr
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Gul Simsek
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Eleanor K. Gill
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands (O.S., I.H.A.H., D.J.D.)
| | - Ilkka H.A. Heinonen
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands (O.S., I.H.A.H., D.J.D.)
- Turku PET Centre, University of Turku and Turku University Hospital, Finland (I.H.A.H.)
| | - Francesco Schianchi
- Faculty of Health, Medicine and Life Sciences, Department of Genetics and Cell Biology, Maastricht University, the Netherlands (F.S., J.J.F.P.L.)
| | - Joost J.F.P. Luiken
- Faculty of Health, Medicine and Life Sciences, Department of Genetics and Cell Biology, Maastricht University, the Netherlands (F.S., J.J.F.P.L.)
| | - Dunja Aksentijevic
- Barts and the London Faculty of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, United Kingdom (D.A.)
| | - Dirk J. Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands (O.S., I.H.A.H., D.J.D.)
| | - Bryan C. Dickinson
- Department of Chemistry, University of Chicago, IL (S.-A.A., T.L., B.C.D.)
| | - Sarah De Val
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - John R. Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada (K.G., J.R.U.)
| | - William Fuller
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (W.F.)
| | - Lisa C. Heather
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| |
Collapse
|
2
|
Susilo RJK, Pramudya M, Dewi FRP, Seftiarini W, Hidayati D, Aunurohim A, Lim V, Herdiansyah MA, Hayati A. Adverse Effect of Polystyrene Nanoplastics in Impairing Glucose Metabolism in Liver Injury. Int J Mol Sci 2025; 26:4870. [PMID: 40430012 PMCID: PMC12112329 DOI: 10.3390/ijms26104870] [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: 04/06/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Polystyrene nanoplastics (PS-NPs) are result from the degradation of plastic and have diameters ranging from 1 nm to 100 nm. The objective of this study is to provide information on the adverse effects of PS-NPs with in vitro and in vivo analyses of liver injury. An in vitro study was conducted using confocal microscopy, flow cytometry, and MTT test analysis. An in vivo study was conducted to determine apoptosis levels, glucose metabolism gene expressions, liver enzymes, and liver histology. Data were analyzed using GraphPad Prism software 10.2.1. The in vitro study showed the absorption of PS-NPs in the cell cytoplasm, the percentage of apoptosis, 3t3, and the WiDr cell lines' viability. The in vivo analysis showed that PS-NPs can stimulate liver injuries, such as inducing the elevation of liver enzymes, necrosis, edema, inflammation, and the dilatation of the portal vein diameter. High levels of caspase-3, caspase-9, and Bax were detected, as well as the expression of several genes including PI3K, AKT, PEPCK, GLUT2, and PK. In conclusion, the in vitro analysis showed the detrimental effects of PS-NPs on cells, such as high levels of apoptosis and low cell viability, while the in vivo studies displayed the impairment of liver tissue and disturbances in glucose metabolism regulation.
Collapse
Affiliation(s)
- Raden Joko Kuncoroningrat Susilo
- Nanotechnology Engineering, Faculty of Advanced Technology and Multidiscipline, Universitas Airlangga, Surabaya 60115, Indonesia;
| | - Manikya Pramudya
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya 60115, Indonesia; (M.P.); (F.R.P.D.); (M.A.H.)
| | - Firli Rahmah Primula Dewi
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya 60115, Indonesia; (M.P.); (F.R.P.D.); (M.A.H.)
| | - Windy Seftiarini
- Graduate School of Biotechnology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | - Dewi Hidayati
- Department of Biology, Faculty of Science and Data Analytics, Institut Teknologi Sepuluh Nopember, Surabaya 60111, Indonesia; (D.H.); (A.A.)
| | - Aunurohim Aunurohim
- Department of Biology, Faculty of Science and Data Analytics, Institut Teknologi Sepuluh Nopember, Surabaya 60111, Indonesia; (D.H.); (A.A.)
| | - Vuanghao Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas 13200, Penang, Malaysia;
| | - Mochammad Aqilah Herdiansyah
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya 60115, Indonesia; (M.P.); (F.R.P.D.); (M.A.H.)
| | - Alfiah Hayati
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya 60115, Indonesia; (M.P.); (F.R.P.D.); (M.A.H.)
| |
Collapse
|
3
|
Yu C, Yin W, Li J, Wu F, Wang S, Han Z, Chen H, Yan X, Cui M. Regulatory Effect of Atorvastatin combined with Berberine on PI3K/Akt/FoxO1 Signaling Pathway in Rats with Hyperlipidemia. Xenobiotica 2025:1-14. [PMID: 40355402 DOI: 10.1080/00498254.2025.2503359] [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: 02/27/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
1.Atorvastatin Calcium (AC) is the first line lipid-lowering drug in clinical. Nowadays, the combination of AC and BBR is often used to treat hyperlipidemia in clinical. In order to determine the mechanism, we investigate the regulatory of atorvastatin combined with berberine on PI3K/Akt/FoxO1 signaling pathway in rats with hyperlipidemia.2.The hyperlipidemia rat model was constructed. Meanwhile, lipid-lowering and liver protective effects were determined by oil red O and H&E method. The expression of PI3K, Akt and FoxO1 was examined by IHC, WB and RT-pCR. The level of CK and LDH in serum was examined by ELISA.3.The results showed that the expression of PI3K, AKT increased and FoxO1 decreased in MC group compared with NC group (P < 0.01). The expression of PI3K, AKT decreased and FoxO1 increased compared with MC group (P < 0.05). The expression of FoxO1 in combination group is lower than AC group. The levels of CK and LDH in AC group increased compared with NC group (P < 0.01), but decreased significantly in AC + BBR group compared with AC group(P < 0.01).4.The combination of AC and BBR could regulate the lipid level by mediating PI3K/Akt/FoxO1, which providing a new references for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Chao Yu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Weihong Yin
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Jiao Li
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Fan Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Siwen Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Zhaoyang Han
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Haoliang Chen
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xueying Yan
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Mingyu Cui
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| |
Collapse
|
4
|
Datta S, Bhattacharjee S, Ghosh S, Ghosh AJ, Saha T, Sen A. Validating the antidiabetic potential of Nakima (Tupistra clarkei Hook.f.), a traditional food from eastern Himalayan region, through network pharmacology and in vivo experimentation. J Pharm Pharmacol 2025:rgaf014. [PMID: 40329835 DOI: 10.1093/jpp/rgaf014] [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: 01/11/2025] [Accepted: 03/25/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To explore and understand the antidiabetic activity of Tupistra clarkei Hook.f. inflorescence, providing a scientific explanation to the ethnomedicinal properties. METHODS The constituents of the plant were determined through GC-MS analysis, which were used for target prediction and network pharmacology to understand how the plant regulates hyperglycaemia and other diabetes complications. These properties were validated in vivo along with further assessment of the antioxidant potential of the plant, both in vitro and in vivo. KEY FINDINGS The plant showed good phenol-flavonoid content, and antioxidant potential both in vitro and in vivo. GC-MS analysis identified 24 constituents of the plant. In silico analysis showed their ability to target 166 proteins that are associated with pathways in controlling hyperglycaemia and other diabetic consequences, protection of pancreatic tissue, insulin secretion, and insulin resistance. This was reflected in the in vivo experiment where T. clarkei showed ability to reduce FBG, LDL-C, VLDL-C levels, improve the levels of HDL-C, and also facilitate reversal of damage in pancreatic islets. CONCLUSION Our study validated the antidiabetic potential Tupistra clarkei inflorescence in the in silico and in vivo assessment, and has proved to have good antioxidant activity and potential against diabetes. However, further clinical trials are essential.
Collapse
Affiliation(s)
- Sutapa Datta
- Molecular Genetics Laboratory, Department of Botany, University of North Bengal, Raja Rammohanpur, Siliguri-734013, India
| | - Soumita Bhattacharjee
- Molecular Genetics Laboratory, Department of Botany, University of North Bengal, Raja Rammohanpur, Siliguri-734013, India
| | - Supriyo Ghosh
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Siliguri-734013, India
| | - Amlan Jyoti Ghosh
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Siliguri-734013, India
| | - Tilak Saha
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Siliguri-734013, India
| | - Arnab Sen
- Molecular Genetics Laboratory, Department of Botany, University of North Bengal, Raja Rammohanpur, Siliguri-734013, India
- Bioinformatics Facility, University of North Bengal, Raja Rammohanpur, Siliguri-734013, India
- Biswa Bangla Genome Centre, University of North Bengal, Raja Rammohanpur, Siliguri-734013, India
| |
Collapse
|
5
|
Panwar A, Malik SO, Adib M, Lopaschuk GD. Cardiac energy metabolism in diabetes: emerging therapeutic targets and clinical implications. Am J Physiol Heart Circ Physiol 2025; 328:H1089-H1112. [PMID: 40192025 DOI: 10.1152/ajpheart.00615.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/15/2024] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Patients with diabetes are at an increased risk for developing diabetic cardiomyopathy and other cardiovascular complications. Alterations in cardiac energy metabolism in patients with diabetes, including an increase in mitochondrial fatty acid oxidation and a decrease in glucose oxidation, are important contributing factors to this increase in cardiovascular disease. A switch from glucose oxidation to fatty acid oxidation not only decreases cardiac efficiency due to increased oxygen consumption but it can also increase reactive oxygen species production, increase lipotoxicity, and redirect glucose into other metabolic pathways that, combined, can lead to heart dysfunction. Currently, there is a lack of therapeutics available to treat diabetes-induced heart failure that specifically target cardiac energy metabolism. However, it is becoming apparent that part of the benefit of existing agents such as GLP-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors may be related to their effects on cardiac energy metabolism. In addition, direct approaches aimed at inhibiting cardiac fatty acid oxidation or increasing glucose oxidation hold future promise as potential therapeutic approaches to treat diabetes-induced cardiovascular disease.
Collapse
Affiliation(s)
- Archee Panwar
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sufyan O Malik
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Muhtasim Adib
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Zhang D, Liang Q, Jiang J, Liu W, Chu Y, Chen Z, Li B, Chen T, Tsao JR, Hu K. SIRT3 mitigates dry eye disease through the activation of autophagy by deacetylation of FOXO1. Exp Eye Res 2025; 254:110328. [PMID: 40064414 DOI: 10.1016/j.exer.2025.110328] [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: 11/24/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
Dry eye disease (DED) is a complex ocular condition characterized by oxidative stress, inflammation, and apoptosis. An increasing number of studies suggest that Sirtuin3 (SIRT3), a mitochondrial deacetylase, may offer protection against related pathologies. Despite these indications, the precise function and underlying mechanisms of SIRT3 in the context of DED have not been fully elucidated. Here, we observed a decline in SIRT3 expression in human corneal epithelial cells (HCE-Ts) and the corneal conjunctiva of mice as the disease advanced. Overexpression of SIRT3 in HCE-Ts reduced the accumulation of reactive oxygen species (ROS), inflammatory cytokines, and the rate of apoptosis, while its inhibition had the opposite effect. Importantly, the function of SIRT3 was exerted through the enhancement of autophagic flux. Further studies have shown that chloroquine-induced inhibition of autophagy neutralized the beneficial effects of SIRT3. In our in vivo experiments, the application of eye drops containing a SIRT3 agonist ameliorated the symptoms of DED and increased corneal autophagy in mice. Mechanistically, our study identified that the deacetylation and nuclear translocation of FOXO1 (Forkhead box O1) are pivotal for the SIRT3-mediated enhancement of autophagic flux. These findings posit that SIRT3 as an encouraging therapeutic target for DED, offering new insights into the disease's underlying mechanisms.
Collapse
Affiliation(s)
- Di Zhang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Qi Liang
- Department of Ophthalmology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, Zhejiang China
| | - Jiaxuan Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Wei Liu
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, China
| | - Yiran Chu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Zeying Chen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Boda Li
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, China
| | - Taige Chen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Jia-Ruei Tsao
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China.
| |
Collapse
|
7
|
Liu JX, Zhang YK, Zhan W, Xie JH, Xu QH, Zhang J, Tai X. USP11 promotes autophagy to attenuate LPS-induced oxidative stress in lung epithelial cells by stabilizing FOXO1 levels. Biochem Biophys Res Commun 2025; 751:151368. [PMID: 39908907 DOI: 10.1016/j.bbrc.2025.151368] [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: 10/03/2024] [Revised: 01/09/2025] [Accepted: 01/19/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Acute lung injury (ALI) is a critical condition characterized by severe inflammation and oxidative stress, leading to high morbidity and mortality. Despite advances in understanding ALI pathophysiology, effective treatment options remain limited. The increasing global burden of ALI, driven by factors such as infections, trauma, and environmental pollutants, emphasizes the urgent need for new therapeutic strategies. This study investigates the role of ubiquitin-specific protease 11 (USP11) in modulating Forkhead box protein O1 (FOXO1) to promote autophagy and alleviate oxidative stress in lung epithelial cells, which could provide novel insights into ALI therapeutic strategies. MATERIALS AND METHODS Bioinformatics were utilized to analyze the expression pattern of USP11 and FOXO1 in ALI, and their functions were detected based on gain- and loss-of function studies in vitro and in vivo. Besides, the effects of USP11 on FOXO1 stability and autophagy were examined through Western blot, immunofluorescence, and co-immunoprecipitation assays. RESULTS USP11 was found to be significantly downregulated in ALI, and its over-expression stabilized FOXO1, enhancing autophagy in lung epithelial cells. USP11 over-expression reduced oxidative stress and inflammatory cytokine production in vitro and in vivo. These results highlight the protective role of the USP11-FOXO1 axis in mitigating ALI pathophysiology. CONCLUSIONS This study identifies USP11 as a key regulator of FOXO1 and autophagy in ALI. The stabilization of FOXO1 through USP11 represents a promising therapeutic strategy for reducing oxidative stress and inflammation in ALI, warranting further clinical investigation.
Collapse
Affiliation(s)
- Jia-Xing Liu
- The First Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yu-Kai Zhang
- The Department of Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Wei Zhan
- The Third Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Jun-Hao Xie
- The Department of Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Qi-Hong Xu
- The Department of Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Jing Zhang
- The First Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Xiang Tai
- The Department of Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.
| |
Collapse
|
8
|
Kielbowski K, Bratborska AW, Bakinowska E, Pawlik A. Sirtuins as therapeutic targets in diabetes. Expert Opin Ther Targets 2025; 29:117-135. [PMID: 40116767 DOI: 10.1080/14728222.2025.2482563] [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: 08/28/2024] [Revised: 03/01/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION Sirtuins (SIRTs) are NAD+-dependent deacetylases that mediate post-translational modifications of proteins. Seven members of the SIRT family have been identified in mammals. Importantly, SIRTs interact with numerous metabolic and inflammatory pathways. Thus, researchers have investigated their role in metabolic and inflammatory disorders. AREAS COVERED In this review, we comprehensively discuss the involvement of SIRTs in the processes of pancreatic β-cell dysfunction, glucose tolerance, insulin secretion, lipid metabolism, and adipocyte functions. In addition, we describe the current evidence regarding modulation of the expression and activity of SIRTs in diabetes, diabetic complications, and obesity. EXPERT OPINION The development of specific SIRT activators and inhibitors that exhibit high selectivity toward specific SIRT isoforms remains a major challenge. This involves the need to elucidate the physiological pathways involving SIRTs, as well as their important role in the development of metabolic disorders. Molecular modeling techniques will be helpful to develop new compounds that modulate the activity of SIRTs, which may contribute to the preparation of new drugs that selectively target specific SIRTs. SIRTs hold promise as potential targets in metabolic disease, but there is much to learn about specific modulators and the final answers will await clinical trials.
Collapse
Affiliation(s)
- Kajetan Kielbowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
9
|
Gupta S, Afzal M, Agrawal N, Almalki WH, Rana M, Gangola S, Chinni SV, Kumar K B, Ali H, Singh SK, Jha SK, Gupta G. Harnessing the FOXO-SIRT1 axis: insights into cellular stress, metabolism, and aging. Biogerontology 2025; 26:65. [PMID: 40011269 DOI: 10.1007/s10522-025-10207-0] [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: 02/02/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
Aging and metabolic disorders share intricate molecular pathways, with the Forkhead box O (FOXO)- Sirtuin 1 (SIRT1) axis emerging as a pivotal regulator of cellular stress adaptation, metabolic homeostasis, and longevity. This axis integrates nutrient signaling with oxidative stress defence, modulating glucose and lipid metabolism, mitochondrial function, and autophagy to maintain cellular stability. FOXO transcription factors, regulated by SIRT1 deacetylation, enhance antioxidant defence mechanisms, activating genes such as superoxide dismutase (SOD) and catalase, thereby counteracting oxidative stress and metabolic dysregulation. Recent evidence highlights the dynamic role of reactive oxygen species (ROS) as secondary messengers in redox signaling, influencing FOXO-SIRT1 activity in metabolic adaptation. Additionally, key redox-sensitive regulators such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) interact with this pathway, orchestrating mitochondrial biogenesis and adaptive stress responses. Pharmacological interventions, including alpha-lipoic acid (ALA), resveratrol, curcumin and NAD+ precursors, exhibit therapeutic potential by enhancing insulin sensitivity, reducing oxidative burden, and restoring metabolic balance. This review synthesizes current advancements in FOXO-SIRT1 regulation, its emerging role in redox homeostasis, and its therapeutic relevance, offering insights into future strategies for combating metabolic dysfunction and aging-related diseases.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Pharmacology, Chameli Devi Institute of Pharmacy, Khandwa Road, Village Umrikheda, Near Tollbooth, Indore, Madhya Pradesh, 452020, India
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gangola
- Department of Microbiology, Graphic Era Deemed to be University, Dehradun, 248002, India
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia
| | - Benod Kumar K
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, New Delhi, India
- Centre for Himalayan Studies, University of Delhi, Delhi, 110007, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
10
|
Chen L, Wang C, Qin L, Zhang H. Parkinson's disease and glucose metabolism impairment. Transl Neurodegener 2025; 14:10. [PMID: 39962629 PMCID: PMC11831814 DOI: 10.1186/s40035-025-00467-8] [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: 07/27/2024] [Accepted: 01/02/2025] [Indexed: 02/21/2025] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. PD patients exhibit varying degrees of abnormal glucose metabolism throughout disease stages. Abnormal glucose metabolism is closely linked to the PD pathogenesis and progression. Key glucose metabolism processes involved in PD include glucose transport, glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation, the pentose phosphate pathway, and gluconeogenesis. Recent studies suggest that glucose metabolism is a potential therapeutic target for PD. In this review, we explore the connection between PD and abnormal glucose metabolism, focusing on the underlying pathophysiological mechanisms. We also summarize potential therapeutic drugs related to glucose metabolism based on results from current cellular and animal model studies.
Collapse
Affiliation(s)
- Liangjing Chen
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chunyu Wang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lixia Qin
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Hainan Zhang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
11
|
Oguh AU, Haemmerle MW, Sen S, Rozo AV, Shrestha S, Cartailler JP, Fazelinia H, Ding H, Preza S, Yang J, Yang X, Sussel L, Alvarez-Dominguez JR, Doliba N, Spruce LA, Arrojo E Drigo R, Stoffers DA. E3 ligase substrate adaptor SPOP fine-tunes the UPR of pancreatic β cells. Genes Dev 2025; 39:261-279. [PMID: 39797759 PMCID: PMC11789638 DOI: 10.1101/gad.352010.124] [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: 06/10/2024] [Accepted: 11/07/2024] [Indexed: 01/13/2025]
Abstract
The Cullin-3 E3 ligase adaptor protein SPOP targets proteins for ubiquitination and proteasomal degradation. We previously established the β-cell transcription factor (TF) and human diabetes gene PDX1 as an SPOP substrate, suggesting a functional role for SPOP in the β cell. Here, we generated a β-cell-specific Spop deletion mouse strain (Spop βKO) and found that Spop is necessary to prevent aberrant basal insulin secretion and for maintaining glucose-stimulated insulin secretion through impacts on glycolysis and glucose-stimulated calcium flux. Integration of proteomic, TF-regulatory gene network, and biochemical analyses identified XBP1 as a functionally important SPOP substrate in pancreatic β cells. Furthermore, loss of SPOP strengthened the IRE1α-XBP1 axis of unfolded protein response (UPR) signaling. ER stress promoted proteasomal degradation of SPOP, supporting a model whereby SPOP fine-tunes XBP1 activation during the UPR. These results position SPOP as a regulator of β-cell function and proper UPR activation.
Collapse
Affiliation(s)
- Alexis U Oguh
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Matthew W Haemmerle
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Sabyasachi Sen
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Andrea V Rozo
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Shristi Shrestha
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Jean-Philippe Cartailler
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Hossein Fazelinia
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Hua Ding
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Sam Preza
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Juxiang Yang
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Xiaodun Yang
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Lori Sussel
- Department of Pediatrics and Cell and Developmental Biology, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Juan R Alvarez-Dominguez
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Nicolai Doliba
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Lynn A Spruce
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Rafael Arrojo E Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Doris A Stoffers
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA;
| |
Collapse
|
12
|
Bahrami P, Aromolaran KA, Aromolaran AS. Proarrhythmic Lipid Inflammatory Mediators: Mechanisms in Obesity Arrhythmias. J Cell Physiol 2025; 240:e70012. [PMID: 39943721 PMCID: PMC11822244 DOI: 10.1002/jcp.70012] [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: 12/08/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025]
Abstract
The prevalence of obesity and associated metabolic disorders such as diabetes is rapidly increasing; therefore, concerns regarding their cardiovascular consequences, including cardiac arrhythmias, are rising. As obesity progresses, the excessively produced lipids accumulate in unconventional areas such as the epicardial adipose tissue (EAT) around the myocardium. Metabolic alterations in obesity contribute to the transformation of these ectopic fat deposits into arrhythmogenic substrates. However, despite advances in therapeutic approaches, particularly in lowering EAT volume and thickness through sodium-glucose co-transporter-2 (SGLT2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists, obese and diabetic patients still suffer from fatal arrhythmias that may lead to sudden cardiac death. Therefore, an investigation into how unappreciated underlying pathways such as lipid mediators contribute to the transformation of adipose tissues into proinflammatory and arrhythmogenic substrates is of significance. Leukotriene B4 (LTB4) is an eicosanoid derived from arachidonic acid and acts as a lipid mediator. LTB4 has recently been identified to be associated with cardiac ion channel modulations and arrhythmogenic conditions in diabetes. LTB4 increases circulatory free fatty acids (FFAs) and has been associated with adipocyte hypertrophy. LTB4 also interferes with insulin signaling pathways, instigating insulin resistance (IR). In addition, LTB4, as a potent chemoattractant, contributes to the mobilization of circulatory immune cells such as monocytes and promotes inflammatory macrophage polarization and macrophage dysfunction. Thus, this review provides a comprehensive overview of LTB4's underlying pathways in obesity; illustrates how these pathways might lead to alterations in cardiac ion channels, currents, and cardiac arrhythmias; and shows how they might pose a therapeutic target for metabolic-associated arrhythmias.
Collapse
Affiliation(s)
- Pegah Bahrami
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI)University of Utah School of MedicineSalt Lake CityUtahUSA
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI)University of Utah School of MedicineSalt Lake CityUtahUSA
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI)University of Utah School of MedicineSalt Lake CityUtahUSA
- Division of Cardiothoracic Surgery, Department of Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine ProgramUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| |
Collapse
|
13
|
Zhou J, Shi Y, Zhao L, Wang R, Luo L, Yin Z. γ-Glutamylcysteine restores glucolipotoxicity-induced islet β-cell apoptosis and dysfunction via inhibiting endoplasmic reticulum stress. Toxicol Appl Pharmacol 2025; 495:117206. [PMID: 39701215 DOI: 10.1016/j.taap.2024.117206] [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: 09/25/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE The impaired function of islet β-cell is associated with the pathogenesis of type 2 diabetes mellitus (T2DM). γ-glutamylcysteine (γ-GC), an immediate precursor of glutathione (GSH), has antioxidant and neuroprotective functions. Its level has been reported to be down-regulated in hyperglycemia. However, whether γ-GC has a protective effect on islet β-cell dysfunction remains elusive. Recently, we explore the molecular mechanism by which γ-GC protects islet β-cell from glucolipotoxicity-induced dysfunction. METHODS In vivo mice models and in vitro cell models were established to examine the therapeutic effects and molecular mechanisms of γ-GC. RESULTS db mice develop impaired glucose-stimulated insulin secretion (GSIS) due to reduced islet number and damaged islet microstructure. Serious oxidative damage, apoptosis and lipid accumulation are also observed in β-cell stimulated by glucolipotoxicity. Mechanistic studies suggest that glucolipotoxicity inhibits PDX-1 nuclear translocation by inducing endoplasmic reticulum (ER) stress, which leads to impaired insulin (INS) secretion in β-cell. Nevertheless, γ-GC as an inhibitor of ER stress can alleviate the damage of islet microstructure in db mice. Importantly, γ-GC promotes INS gene expression and GSIS through driving nuclear translocation of PDX-1, thereby enhancing intracellular INS content. Moreover, treatment with γ-GC can also mitigate oxidative damage, apoptosis and lipid accumulation of β-cell, resulting in ameliorating islet β-cell dysfunction induced by glucolipotoxicity. CONCLUSION Our results support the use of γ-GC as an inhibitor of ER stress for prevention and treatment of T2DM in the future.
Collapse
Affiliation(s)
- Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Yingying Shi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lishuang Zhao
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Rong Wang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
14
|
Golounina O, Minniakhmetov I, Salakhov R, Khusainova R, Zakharova E, Bychkov I, Mokrysheva N. Pathogenetic therapeutic approaches for endocrine diseases based on antisense oligonucleotides and RNA-interference. Front Endocrinol (Lausanne) 2025; 16:1525373. [PMID: 39944202 PMCID: PMC11813780 DOI: 10.3389/fendo.2025.1525373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/13/2025] [Indexed: 05/09/2025] Open
Abstract
Molecular therapy uses nucleic acid-based therapeutics agents and becomes a promising alternative for disease conditions unresponsive to traditional pharmaceutical approaches. Antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs) are two well-known strategies used to modulate gene expression. RNA-targeted therapy can precisely modulate the function of target RNA with minimal off-target effects and can be rationally designed based on sequence data. ASOs and siRNA-based drugs have unique capabilities for using in target groups of patients or can be tailored as patient-customized N-of-1 therapeutic approach. Antisense therapy can be utilized not only for the treatment of monogenic diseases but also holds significant promise for addressing polygenic and complex diseases by targeting key genes and molecular pathways involved in disease pathogenesis. In the context of endocrine disorders, molecular therapy is particularly effective in modulating pathogenic mechanisms such as defective insulin signaling, beta-cell dysfunction and hormonal imbalances. Furthermore, siRNA and ASOs have the ability to downregulate overactive signaling pathways that contribute to complex, non-monogenic endocrine disorders, thereby addressing these conditions at their molecular origin. ASOs are also being studied worldwide as unique candidates for developing therapies for N-of-1 therapies. The sequence-specific ASOs binding provides exceptional accuracy in N-of-1 approaches, when the oligonucleotide can be targeted to a patient's exact mutant sequence. In this review we focus on diseases of the endocrine system and discuss potential RNA-targeted therapeutic opportunities in diabetes mellitus, including monogenic beta cell diabetes, and obesity, including syndrome obesity and monogenic obesity, as well as in non-monogenic or complex endocrine disorders. We also provide an overview of currently developed and available antisense molecules, and describe potentials of antisense-based therapeutics for the treatment of rare and «ultrarare» endocrine diseases.
Collapse
Affiliation(s)
- Olga Golounina
- Department of Clinical Endocrinology, Endocrinology Research Centre, Moscow, Russia
| | - Ildar Minniakhmetov
- Laboratory of Genomic Medicine, Endocrinology Research Centre, Moscow, Russia
| | - Ramil Salakhov
- Laboratory of Genomic Medicine, Endocrinology Research Centre, Moscow, Russia
| | - Rita Khusainova
- Laboratory of Genomic Medicine, Endocrinology Research Centre, Moscow, Russia
| | - Ekaterina Zakharova
- Selective Screening Laboratory, Research Centre for Medical Genetics, Moscow, Russia
| | - Igor Bychkov
- Laboratory of Experimental Gene Therapy for Inherited Metabolic Diseases, Research Centre for Medical Genetics, Moscow, Russia
| | - Natalia Mokrysheva
- Department of Clinical Endocrinology, Endocrinology Research Centre, Moscow, Russia
| |
Collapse
|
15
|
Kiruba B, Naidu A, Sundararajan V, Lulu S S. Mapping integral cell-type-specific interferon-induced gene regulatory networks (GRNs) involved in systemic lupus erythematosus using systems and computational analysis. Heliyon 2025; 11:e41342. [PMID: 39844998 PMCID: PMC11751531 DOI: 10.1016/j.heliyon.2024.e41342] [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] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disorder characterized by the production of autoantibodies, resulting in inflammation and organ damage. Although extensive research has been conducted on SLE pathogenesis, a comprehensive understanding of its molecular landscape in different cell types has not been achieved. This study uncovers the molecular mechanisms of the disease by thoroughly examining gene regulatory networks within neutrophils, dendritic cells, T cells, and B cells. Firstly, we identified genes and ncRNAs with differential expression in SLE patients compared to controls for different cell types. Furthermore, the derived differentially expressed genes were curated based on immune functions using functional enrichment analysis to create a protein-protein interaction network. Topological network analysis of the formed genes revealed key hub genes associated with each of the cell types. To understand the regulatory mechanism through which these hub genes function in the diseased state, their associations with transcription factors, and non-coding RNAs in different immune cell types were investigated through correlation analysis and regression models. Finally, by integrating these findings, distinct gene regulatory networks were constructed, which provide a novel perspective on the molecular, cellular, and immunological landscapes of SLE. Importantly, we reveal the crucial role of IRF3, IRF7, and STAT1 in neutrophils, dendritic cells, and T cells, where their aberrant upregulation in disease states might enhance the production of type I IFN. Furthermore, we found MYB to be a crucial regulator that might activate T cells toward autoimmune responses in SLE. Similarly, in B-cell lymphocytes, we found FOXO1 to be a key player in autophagy and chemokine regulation. These findings were also validated using single-cell RNASeq analysis using an independent dataset. Genotype variations of these genes were also explored using the GWAS central database to ensure their targetability. These findings indicate that IRF3, IRF7, STAT1, MYB, and FOXO1 are promising targets for therapeutic interventions for SLE.
Collapse
Affiliation(s)
- Blessy Kiruba
- Department of Biosciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| | - Akshayata Naidu
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| | - Vino Sundararajan
- Department of Biosciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| | - Sajitha Lulu S
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| |
Collapse
|
16
|
Li H, Zou L, Long Z, Zhan J. Immunometabolic alterations in type 2 diabetes mellitus revealed by single-cell RNA sequencing: insights into subtypes and therapeutic targets. Front Immunol 2025; 15:1537909. [PMID: 39877357 PMCID: PMC11772204 DOI: 10.3389/fimmu.2024.1537909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Background Type 2 Diabetes Mellitus (T2DM) represents a major global health challenge, marked by chronic hyperglycemia, insulin resistance, and immune system dysfunction. Immune cells, including T cells and monocytes, play a pivotal role in driving systemic inflammation in T2DM; however, the underlying single-cell mechanisms remain inadequately defined. Methods Single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) from 37 patients with T2DM and 11 healthy controls (HC) was conducted. Immune cell types were identified through clustering analysis, followed by differential expression and pathway analysis. Metabolic heterogeneity within T cell subpopulations was evaluated using Gene Set Variation Analysis (GSVA). Machine learning models were constructed to classify T2DM subtypes based on metabolic signatures, and T-cell-monocyte interactions were explored to assess immune crosstalk. Transcription factor (TF) activity was analyzed, and drug enrichment analysis was performed to identify potential therapeutic targets. Results In patients with T2DM, a marked increase in monocytes and a decrease in CD4+ T cells were observed, indicating immune dysregulation. Significant metabolic diversity within T cell subpopulations led to the classification of patients with T2DM into three distinct subtypes (A-C), with HC grouped as D. Enhanced intercellular communication, particularly through the MHC-I pathway, was evident in T2DM subtypes. Machine learning models effectively classified T2DM subtypes based on metabolic signatures, achieving an AUC > 0.84. Analysis of TF activity identified pivotal regulators, including NF-kB, STAT3, and FOXO1, associated with immune and metabolic disturbances in T2DM. Drug enrichment analysis highlighted potential therapeutic agents targeting these TFs and related pathways, including Suloctidil, Chlorpropamide, and other compounds modulating inflammatory and metabolic pathways. Conclusion This study underscores significant immunometabolic dysfunction in T2DM, characterized by alterations in immune cell composition, metabolic pathways, and intercellular communication. The identification of critical TFs and the development of drug enrichment profiles highlight the potential for personalized therapeutic strategies, emphasizing the need for integrated immunological and metabolic approaches in T2DM management.
Collapse
Affiliation(s)
- Huahua Li
- Department of Geriatric, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Lingling Zou
- Department of Geriatric, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhaowei Long
- Department of Geriatric, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Junkun Zhan
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, China
| |
Collapse
|
17
|
Shi JH, Shi YB, Qiu ST, Song Y. Role of the Transcription Factor FoxO in Type 2 Diabetes and Its Complications. Crit Rev Eukaryot Gene Expr 2025; 35:85-103. [PMID: 39964972 DOI: 10.1615/critreveukaryotgeneexpr.2025057309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
FoxO proteins represent a subfamily of the forkhead box family (Fox) superfamily of proteins. It is involved in cell proliferation, differentiation, oxidative stress, apoptosis as well as tumors and metabolic disorders by regulating cellular functions. This paper aims to summarize the role of the transcription factor FoxO in type 2 diabetes and its complications, which may add to the potential of FoxO as a therapeutic target for future research. The transcription factor FoxO is expressed in various tissues and participates in various bodily functions including cell proliferation, differentiation, apoptosis, tumor therapy, and metabolic processes, playing a crucial role in the human body. FoxO plays a positive role in attenuating oxidative stress, inflammation, and metabolic disorders, which are the main causes of type 2 diabetes and its complications. FoxO plays an important role in the regulation of type 2 diabetes and its complications, and more precise targeting studies of FoxO will help to prevent, regulate, and treat diabetes-related diseases.
Collapse
Affiliation(s)
- Jing Hui Shi
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yi Biao Shi
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Si Tian Qiu
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, 18# Chaowang Road, Hangzhou City, Zhejiang Province 310014, P.R. China
| |
Collapse
|
18
|
Yang Y, Huang B, Qin Y, Wang D, Jin Y, Su L, Wang Q, Pan Y, Zhang Y, Shen Y, Hu W, Cao Z, Jin L, Zhang F. Adipocyte microRNA-802 promotes adipose tissue inflammation and insulin resistance by modulating macrophages in obesity. eLife 2024; 13:e99162. [PMID: 39589393 DOI: 10.7554/elife.99162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024] Open
Abstract
Adipose tissue inflammation is now considered to be a key process underlying metabolic diseases in obese individuals. However, it remains unclear how adipose inflammation is initiated and maintained or the mechanism by which inflammation develops. We found that microRNA-802 (Mir802) expression in adipose tissue is progressively increased with the development of dietary obesity in obese mice and humans. The increasing trend of Mir802 preceded the accumulation of macrophages. Adipose tissue-specific knockout of Mir802 lowered macrophage infiltration and ameliorated systemic insulin resistance. Conversely, the specific overexpression of Mir802 in adipose tissue aggravated adipose inflammation in mice fed a high-fat diet. Mechanistically, Mir802 activates noncanonical and canonical NF-κB pathways by targeting its negative regulator, TRAF3. Next, NF-κB orchestrated the expression of chemokines and SREBP1, leading to strong recruitment and M1-like polarization of macrophages. Our findings indicate that Mir802 endows adipose tissue with the ability to recruit and polarize macrophages, which underscores Mir802 as an innovative and attractive candidate for miRNA-based immune therapy for adipose inflammation.
Collapse
Affiliation(s)
- Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Bin Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yimeng Qin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Danwei Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yinuo Jin
- NanJing HanKai Academy, Nanjing, China
| | - Linmin Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qingxin Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yumeng Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenjun Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhengyu Cao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangfang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
19
|
Hlavac K, Pavelkova P, Ondrisova L, Mraz M. FoxO1 signaling in B cell malignancies and its therapeutic targeting. FEBS Lett 2024. [PMID: 39533662 DOI: 10.1002/1873-3468.15057] [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: 07/16/2024] [Revised: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
FoxO transcription factors (FoxO1, FoxO3a, FoxO4, FoxO6) are a highly evolutionary conserved subfamily of the 'forkhead' box proteins. They have traditionally been considered tumor suppressors, but FoxO1 also exhibits oncogenic properties. The complex nature of FoxO1 is illustrated by its various roles in B cell development and differentiation, immunoglobulin gene rearrangement and cell-surface B cell receptor (BCR) structure, DNA damage control, cell cycle regulation, and germinal center reaction. FoxO1 is tightly regulated at a transcriptional (STAT3, HEB, EBF, FoxOs) and post-transcriptional level (Akt, AMPK, CDK2, GSK3, IKKs, JNK, MAPK/Erk, SGK1, miRNA). In B cell malignancies, recurrent FoxO1 activating mutations (S22/T24) and aberrant nuclear export and activity have been described, underscoring the potential of its therapeutic inhibition. Here, we review FoxO1's roles across B cell and myeloid malignancies, namely acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Burkitt lymphoma (BL), Hodgkin lymphoma (HL), and multiple myeloma (MM). We also discuss preclinical evidence for FoxO1 targeting by currently available inhibitors (AS1708727, AS1842856, cpd10).
Collapse
Affiliation(s)
- Krystof Hlavac
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Petra Pavelkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Laura Ondrisova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Marek Mraz
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| |
Collapse
|
20
|
Damarov IS, Korbolina EE, Rykova EY, Merkulova TI. Multi-Omics Analysis Revealed the rSNPs Potentially Involved in T2DM Pathogenic Mechanism and Metformin Response. Int J Mol Sci 2024; 25:9297. [PMID: 39273245 PMCID: PMC11394919 DOI: 10.3390/ijms25179297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The goal of our study was to identify and assess the functionally significant SNPs with potentially important roles in the development of type 2 diabetes mellitus (T2DM) and/or their effect on individual response to antihyperglycemic medication with metformin. We applied a bioinformatics approach to identify the regulatory SNPs (rSNPs) associated with allele-asymmetric binding and expression events in our paired ChIP-seq and RNA-seq data for peripheral blood mononuclear cells (PBMCs) of nine healthy individuals. The rSNP outcomes were analyzed using public data from the GWAS (Genome-Wide Association Studies) and Genotype-Tissue Expression (GTEx). The differentially expressed genes (DEGs) between healthy and T2DM individuals (GSE221521), including metformin responders and non-responders (GSE153315), were searched for in GEO RNA-seq data. The DEGs harboring rSNPs were analyzed using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We identified 14,796 rSNPs in the promoters of 5132 genes of human PBMCs. We found 4280 rSNPs to associate with both phenotypic traits (GWAS) and expression quantitative trait loci (eQTLs) from GTEx. Between T2DM patients and controls, 3810 rSNPs were detected in the promoters of 1284 DEGs. Based on the protein-protein interaction (PPI) network, we identified 31 upregulated hub genes, including the genes involved in inflammation, obesity, and insulin resistance. The top-ranked 10 enriched KEGG pathways for these hubs included insulin, AMPK, and FoxO signaling pathways. Between metformin responders and non-responders, 367 rSNPs were found in the promoters of 131 DEGs. Genes encoding transcription factors and transcription regulators were the most widely represented group and many were shown to be involved in the T2DM pathogenesis. We have formed a list of human rSNPs that add functional interpretation to the T2DM-association signals identified in GWAS. The results suggest candidate causal regulatory variants for T2DM, with strong enrichment in the pathways related to glucose metabolism, inflammation, and the effects of metformin.
Collapse
Affiliation(s)
- Igor S Damarov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Elena E Korbolina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Elena Y Rykova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Engineering Problems of Ecology, Novosibirsk State Technical University, 630087 Novosibirsk, Russia
| | - Tatiana I Merkulova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
21
|
Machi JF, Altilio I, Qi Y, Morales AA, Silvestre DH, Hernandez DR, Da Costa-Santos N, Santana AG, Neghabi M, Nategh P, Castro TL, Werneck-de-Castro JP, Ranji M, Evangelista FS, Vazquez-Padron RI, Bernal-Mizrachi E, Rodrigues CO. Endothelial c-Myc knockout disrupts metabolic homeostasis and triggers the development of obesity. Front Cell Dev Biol 2024; 12:1407097. [PMID: 39100099 PMCID: PMC11294153 DOI: 10.3389/fcell.2024.1407097] [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: 03/27/2024] [Accepted: 06/10/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction: Obesity is a major risk factor associated with multiple pathological conditions including diabetes and cardiovascular disease. Endothelial dysfunction is an early predictor of obesity. However, little is known regarding how early endothelial changes trigger obesity. In the present work we report a novel endothelial-mediated mechanism essential for regulation of metabolic homeostasis, driven by c-Myc. Methods: We used conditional knockout (EC-Myc KO) and overexpression (EC-Myc OE) mouse models to investigate the endothelial-specific role of c-Myc in metabolic homeostasis during aging and high-fat diet exposure. Body weight and metabolic parameters were collected over time and tissue samples collected at endpoint for biochemical, pathology and RNA-sequencing analysis. Animals exposed to high-fat diet were also evaluated for cardiac dysfunction. Results: In the present study we demonstrate that EC-Myc KO triggers endothelial dysfunction, which precedes progressive increase in body weight during aging, under normal dietary conditions. At endpoint, EC-Myc KO animals showed significant increase in white adipose tissue mass relative to control littermates, which was associated with sex-specific changes in whole body metabolism and increase in systemic leptin. Overexpression of endothelial c-Myc attenuated diet-induced obesity and visceral fat accumulation and prevented the development of glucose intolerance and cardiac dysfunction. Transcriptome analysis of skeletal muscle suggests that the protective effects promoted by endothelial c-Myc overexpression are associated with the expression of genes known to increase weight loss, energy expenditure and glucose tolerance. Conclusion: Our results show a novel important role for endothelial c-Myc in regulating metabolic homeostasis and suggests its potential targeting in preventing obesity and associated complications such as diabetes type-2 and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Jacqueline F. Machi
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Isabella Altilio
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Yue Qi
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Alejo A. Morales
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego H. Silvestre
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diana R. Hernandez
- DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Nicolas Da Costa-Santos
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Aline G. Santana
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Mehrnoosh Neghabi
- Department of Electrical Engineering and Computer Science, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Parisa Nategh
- Department of Electrical Engineering and Computer Science, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Thiago L. Castro
- School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - João P. Werneck-de-Castro
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Mahsa Ranji
- Department of Electrical Engineering and Computer Science, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, United States
| | | | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Claudia O. Rodrigues
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
22
|
Sasako T. Exploring mechanisms of insulin action and strategies to treat diabetes. Endocr J 2024; 71:651-660. [PMID: 38811207 DOI: 10.1507/endocrj.ej24-0003] [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] [Indexed: 05/31/2024] Open
Abstract
Insulin is a hormone that positively regulates anabolism and cell growth, whereas diabetes mellitus is a disease characterized by hyperglycemia associated with impaired insulin action. My colleagues and I have elucidated multifaceted insulin action in various tissues mainly by means of model mice. In the liver, insulin regulates endoplasmic reticulum (ER) stress response during feeding, whereas ER stress 'response failure' contributes to the development of steatohepatitis comorbid with diabetes. Not only the liver but also the proximal tubules of the kidney are important in the regulation of gluconeogenesis, and we revealed that insulin suppresses gluconeogenesis in accordance with absorbed glucose in the latter tissue. In skeletal muscle, another important insulin-targeted tissue, impaired insulin/IGF-1 signaling leads not only to sarcopenia, an aging-related disease of skeletal muscle, but also to osteopenia and shorter longevity. Aging is regulated by adipokines as well, and it should be considered that aging could be accelerated by 'imbalanced adipokines' in patients with a genetic background of progeria. Moreover, we reported the effects of intensive multifactorial intervention on diabetic vascular complications and mortality in patients with type 2 diabetes in a large-scale clinical trial, the J-DOIT3, and the results of subsequent sub-analyses of renal events and fracture events. Various approaches of research enable us of endocrinologists to elucidate the physiology of hormone signaling, the mechanisms underlying the development of endocrine diseases, and the appropriate treatment measures. These approaches also raise fundamental questions, but addressing them in an appropriate manner will surely contribute to the further development of endocrinology.
Collapse
Affiliation(s)
- Takayoshi Sasako
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
23
|
Gupta RS, Grover AS, Kumar P, Goel A, Banik SP, Chakraborty S, Rungta M, Bagchi M, Pal P, Bagchi D. A randomized double blind placebo controlled trial to assess the safety and efficacy of a patented fenugreek ( Trigonella foenum-graecum) seed extract in Type 2 diabetics. Food Nutr Res 2024; 68:10667. [PMID: 38863744 PMCID: PMC11165257 DOI: 10.29219/fnr.v68.10667] [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: 03/18/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
Background Fenugreek plant (Trigonella foenum-graecum) constitutes a traditionally acclaimed herbal remedy for many human ailments including diabetes, obesity, neurodegenerative diseases, and reproductive disorders. It is also used as an effective anti-oxidative, anti-inflammatory, antibacterial, and anti-fungal agent. The seed of the plant is especially enriched in several bioactive molecules including polyphenols, saponins, alkaloids, and flavonoids and has demonstrated potential to act as an antidiabetic phytotherapeutic. A novel patented formulation (Fenfuro®) was developed in our laboratory from the fenugreek seeds which contained >45% furostanolic saponins (HPLC). Objective A placebo-controlled clinical compliance study was designed to assess the effects of complementing Fenfuro® on a randomized group of human volunteers on antidiabetic therapy (Metformin and sulphonylurea) in controlling the glycemic index along with simultaneous safety assessment. Study methodology and trial design In a randomized double-blind, placebo-controlled trial, 42 individuals (21 male and 21 female volunteers) in the treatment group (out of 57 enrolled) and 39 individuals (17 male and 22 female volunteers) in the placebo group (out of 47 enrolled), all on antidiabetic therapy with Metformin/Metformin with sulphonyl urea within the age group of 18-65 years were administered either 1,000 mg (500 mg × 2) (Fenfuro®) capsules or placebo over a period of 12 consecutive weeks. Fasting and postprandial glucose along with glycated hemoglobin were determined as primary outcomes to assess the antidiabetic potential of the formulation. Moreover, in order to evaluate the safety of the formulation, C-peptide and Thyroid Stimulating Hormone (TSH) levels as well as immunohematological parameters were assessed between the treatment and placebo groups at the completion of the study. Results After 12 weeks of administration, both fasting as well as postprandial serum glucose levels decreased by 38 and 44% respectively in the treatment group. Simultaneously, a significant reduction in glycated hemoglobin by about 34.7% was also noted. The formulation did not have any adverse effect on the study subjects as there was no significant change in C- peptide level and TSH level; liver, kidney, and cardiovascular function was also found to be normal as assessed by serum levels of key immunohematological parameters. No adverse events were reported. Conclusion This clinical compliance study re-instated and established the safety and efficacy of Fenfuro® as an effective phytotherapeutic to treat hyperglycemia.
Collapse
Affiliation(s)
- Rajinder Singh Gupta
- Department of Medicine, Gian Sagar Medical College & Hospital, Banur, Patiala, India
| | - Amarjit Singh Grover
- Department of Surgery, Gian Sagar Medical College & Hospital, Banur, Patiala, India
| | - Pawan Kumar
- R&D Department, Chemical Resources (CHERESO), Panchkula, Haryana, India
| | - Apurva Goel
- Regulatory Department, Chemical Resources (CHERESO), Panchkula, Haryana, India
| | - Samudra P. Banik
- Department of Microbiology, Maulana Azad College, Kolkata, India
| | - Sanjoy Chakraborty
- Department of Biological Sciences, New York City College of Technology/CUNY, Brooklyn, NY, USA
| | - Mehul Rungta
- R&D Department, Chemical Resources (CHERESO), Panchkula, Haryana, India
| | | | - Partha Pal
- Department of Statistics, Maulana Azad College, Kolkata, India
| | - Debasis Bagchi
- Department of Biology, College of Arts and Sciences, and Department of Psychology, Gordon F. Derner School of Psychology, Adelphi University, Garden City, NY, USA
| |
Collapse
|
24
|
Ge B, Yan K, Sang R, Wang W, Liu X, Yu M, Liu X, Qiu Q, Zhang X. Integrated network toxicology, molecular docking, and in vivo experiments to elucidate molecular mechanism of aflatoxin B1 hepatotoxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 275:116278. [PMID: 38564860 DOI: 10.1016/j.ecoenv.2024.116278] [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: 01/11/2024] [Revised: 03/07/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Due to the rise in temperature and sea level caused by climate change, the detection rate of aflatoxin B1 (AFB1) in food crops has increased dramatically, and the frequency and severity of aflatoxicosis in humans and animals are also increasing. AFB1 has strong hepatotoxicity, causing severe liver damage and even cancer. However, the mechanism of AFB1 hepatotoxicity remains unclear. By integrating network toxicology, molecular docking and in vivo experiments, this research was designed to explore the potential hepatotoxicity mechanisms of AFB1. Thirty-three intersection targets for AFB1-induced liver damage were identified using online databases. PI3K/AKT1, MAPK, FOXO1 signaling pathways, and apoptosis were significantly enriched. In addition, the proteins of ALB, AKT1, PIK3CG, MAPK8, HSP90AA1, PPARA, MAPK1, EGFR, FOXO1, and IGF1 exhibited good affinity with AFB1. In vivo experiments, significant pathological changes occurred in the liver of mice. AFB1 induction increased the expression levels of EGFR, ERK, and FOXO1, and decreased the expression levsls of PI3K and AKT1. Moreover, AFB1 treatment caused an increase in Caspase3 expression, and a decrease in Bcl2/Bax ratio. By combining network toxicology with in vivo experiments, this study confirms for the first time that AFB1 promotes the FOXO1 signaling pathway by inactivating PI3K/AKT1 and activating EGFR/ERK signaling pathways, hence aggravating hepatocyte apoptosis. This research provides new strategies for studying the toxicity of environmental pollutants and new possible targets for the development of hepatoprotective drugs.
Collapse
Affiliation(s)
- Bingjie Ge
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Kexin Yan
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Rui Sang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Wei Wang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Xinman Liu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Minghong Yu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Xiaotong Liu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Qian Qiu
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Xuemei Zhang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China.
| |
Collapse
|
25
|
Huang HYR, Badar S, Said M, Shah S, Bharadwaj HR, Ramamoorthy K, Alrawashdeh MM, Haroon F, Basit J, Saeed S, Aji N, Tse G, Roy P, Bardhan M. The advent of RNA-based therapeutics for metabolic syndrome and associated conditions: a comprehensive review of the literature. Mol Biol Rep 2024; 51:493. [PMID: 38580818 DOI: 10.1007/s11033-024-09457-x] [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: 01/19/2024] [Accepted: 03/18/2024] [Indexed: 04/07/2024]
Abstract
Metabolic syndrome (MetS) is a prevalent and intricate health condition affecting a significant global population, characterized by a cluster of metabolic and hormonal disorders disrupting lipid and glucose metabolism pathways. Clinical manifestations encompass obesity, dyslipidemia, insulin resistance, and hypertension, contributing to heightened risks of diabetes and cardiovascular diseases. Existing medications often fall short in addressing the syndrome's multifaceted nature, leading to suboptimal treatment outcomes and potential long-term health risks. This scenario underscores the pressing need for innovative therapeutic approaches in MetS management. RNA-based treatments, employing small interfering RNAs (siRNAs), microRNAs (miRNAs), and antisense oligonucleotides (ASOs), emerge as promising strategies to target underlying biological abnormalities. However, a summary of research available on the role of RNA-based therapeutics in MetS and related co-morbidities is limited. Murine models and human studies have been separately interrogated to determine whether there have been recent advancements in RNA-based therapeutics to offer a comprehensive understanding of treatment available for MetS. In a narrative fashion, we searched for relevant articles pertaining to MetS co-morbidities such as cardiovascular disease, fatty liver disease, dementia, colorectal cancer, and endocrine abnormalities. We emphasize the urgency of exploring novel therapeutic avenues to address the intricate pathophysiology of MetS and underscore the potential of RNA-based treatments, coupled with advanced delivery systems, as a transformative approach for achieving more comprehensive and efficacious outcomes in MetS patients.
Collapse
Affiliation(s)
- Helen Ye Rim Huang
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sarah Badar
- Department of Biomedical Science, The University of the West Scotland, Paisley, Scotland
| | - Mohammad Said
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Siddiqah Shah
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Krishna Ramamoorthy
- Department of Biochemistry and Microbiology, Rutgers University-New Brunswick, Brunswick, NJ, USA
| | | | | | - Jawad Basit
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Sajeel Saeed
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Narjiss Aji
- Faculty of Medicine and Health, McGill University, Montreal, QC, Canada
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Priyanka Roy
- Directorate of Factories, Department of Labour, Government of West Bengal, Kolkata, India
| | - Mainak Bardhan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA.
| |
Collapse
|
26
|
Jia Y, Yu Y, Gao C, Li Y, Li C, Ding Z, Kong Q, Liu L. Roles of heat shock protein A12A in the development of diabetic cardiomyopathy. Cell Stress Chaperones 2024; 29:272-284. [PMID: 38485044 PMCID: PMC10972809 DOI: 10.1016/j.cstres.2024.03.004] [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: 12/07/2023] [Revised: 02/08/2024] [Accepted: 03/09/2024] [Indexed: 03/24/2024] Open
Abstract
Long-term hyperglycemia can lead to diabetic cardiomyopathy (DCM), a main lethal complication of diabetes. However, the mechanisms underlying DCM development have not been fully elucidated. Heat shock protein A12A (HSPA12A) is the atypic member of the Heat shock 70kDa protein family. In the present study, we found that the expression of HSPA12A was upregulated in the hearts of mice with streptozotocin-induced diabetes, while ablation of HSPA12A improved cardiac systolic and diastolic dysfunction and increased cumulative survival of diabetic mice. An increased expression of HSPA12A was also found in H9c2 cardiac cells following treatment with high glucose (HG), while overexpression of HSPA12A-enhanced the HG-induced cardiac cell death, as reflected by higher levels of propidium iodide cells, lactate dehydrogenase leakage, and caspase 3 cleavage. Moreover, the HG-induced increase of oxidative stress, as indicated by dihydroethidium staining, was exaggerated by HSPA12A overexpression. Further studies demonstrated that the HG-induced increases of protein kinase B and forkhead box transcription factors 1 phosphorylation were diminished by HSPA12A overexpression, while pharmacologically inhibition of protein kinase B further enhanced the HG-induced lactate dehydrogenase leakage in HSPA12A overexpressed cardiac cells. Together, the results suggest that hyperglycemia upregulated HSPA12A expression in cardiac cells, by which induced cell death to promote DCM development. Targeting HSPA12A may serve as a potential approach for DCM management.
Collapse
Affiliation(s)
- Yunxiao Jia
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yunhao Yu
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chenxi Gao
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, TN, USA
| | - Zhengnian Ding
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qiuyue Kong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Li Liu
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
27
|
Sharma C, Hamza A, Boyle E, Donu D, Cen Y. Post-Translational Modifications and Diabetes. Biomolecules 2024; 14:310. [PMID: 38540730 PMCID: PMC10968569 DOI: 10.3390/biom14030310] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 09/22/2024] Open
Abstract
Diabetes and its associated complications have increasingly become major challenges for global healthcare. The current therapeutic strategies involve insulin replacement therapy for type 1 diabetes (T1D) and small-molecule drugs for type 2 diabetes (T2D). Despite these advances, the complex nature of diabetes necessitates innovative clinical interventions for effective treatment and complication prevention. Accumulative evidence suggests that protein post-translational modifications (PTMs), including glycosylation, phosphorylation, acetylation, and SUMOylation, play important roles in diabetes and its pathological consequences. Therefore, the investigation of these PTMs not only sheds important light on the mechanistic regulation of diabetes but also opens new avenues for targeted therapies. Here, we offer a comprehensive overview of the role of several PTMs in diabetes, focusing on the most recent advances in understanding their functions and regulatory mechanisms. Additionally, we summarize the pharmacological interventions targeting PTMs that have advanced into clinical trials for the treatment of diabetes. Current challenges and future perspectives are also provided.
Collapse
Affiliation(s)
- Chiranjeev Sharma
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Abu Hamza
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Emily Boyle
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Dickson Donu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
28
|
Long Y, Mao C, Liu S, Tao Y, Xiao D. Epigenetic modifications in obesity-associated diseases. MedComm (Beijing) 2024; 5:e496. [PMID: 38405061 PMCID: PMC10893559 DOI: 10.1002/mco2.496] [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: 08/03/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
The global prevalence of obesity has reached epidemic levels, significantly elevating the susceptibility to various cardiometabolic conditions and certain types of cancer. In addition to causing metabolic abnormalities such as insulin resistance (IR), elevated blood glucose and lipids, and ectopic fat deposition, obesity can also damage pancreatic islet cells, endothelial cells, and cardiomyocytes through chronic inflammation, and even promote the development of a microenvironment conducive to cancer initiation. Improper dietary habits and lack of physical exercise are important behavioral factors that increase the risk of obesity, which can affect gene expression through epigenetic modifications. Epigenetic alterations can occur in early stage of obesity, some of which are reversible, while others persist over time and lead to obesity-related complications. Therefore, the dynamic adjustability of epigenetic modifications can be leveraged to reverse the development of obesity-associated diseases through behavioral interventions, drugs, and bariatric surgery. This review provides a comprehensive summary of the impact of epigenetic regulation on the initiation and development of obesity-associated cancers, type 2 diabetes, and cardiovascular diseases, establishing a theoretical basis for prevention, diagnosis, and treatment of these conditions.
Collapse
Affiliation(s)
- Yiqian Long
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Chao Mao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaChina
| | - Shuang Liu
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yongguang Tao
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaChina
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Department of Thoracic SurgerySecond Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Desheng Xiao
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, School of Basic MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|