101
|
Arun D, Munir W, Schmitt LV, Vyas R, Ravindran JI, Bashir M, Williams IM, Velayudhan B, Idhrees M. Exploring the Correlation and Protective Role of Diabetes Mellitus in Aortic Aneurysm Disease. Front Cardiovasc Med 2021; 8:769343. [PMID: 34820431 PMCID: PMC8606667 DOI: 10.3389/fcvm.2021.769343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/11/2021] [Indexed: 01/04/2023] Open
Abstract
Introduction: Diabetes mellitus is recognised as a significant risk factor for cardiovascular and peripheral vascular disease, as the abnormal metabolic state increases the risk for atherosclerosis, occlusive arterial disease and vascular dysfunction. There have been reports of potential association across the literature that illustrates a link between diabetes mellitus and aortic aneurysm, with the former having a protective role on the development of the latter. Methods: A thorough literature search was performed through electronic databases, to provide a comprehensive review of the study's reporting on the association of diabetes mellitus and aortic aneurysm, discussing the mechanisms that have been reported; furthemore, we reviewed the reports of the impact of oral hypoglycameic agents on aortic aneurysms. Results: Various proposed mechanisms are involved in this protective process including endothelial dysfunction, chronic hyperglycemia and insulin resistance. The evidence suggests a negative association between these disease process, with prevelance of diabetes mellitus resulting in lower rates of aortic aneurysm, via its protective mechanistic action. The increase in advanced glycation end products, increased arterial stiffness and vascular remodelling seen in diabetes, was found to have a profound impact on aneurysm development, its slow progression and lower rupture rate in these individuals. This review has also highlighted the role of oral hypoglycaemic agents having a protective effect against AA disease. Conclusion: A decrease in development, progression and mortality from aortic aneurysms as well as reduced rates of dissection, have been observed in those with diabetes. This review has provided a comprehensive insight on the effect of diabetes and its physiological processes, and elements of its con-committant treatment, having a protective role against these aortic diseases.
Collapse
Affiliation(s)
- Divyatha Arun
- Department of Endocrinology, Columbia Asia Referral Hospital, A Unit of Manipal Hospital, Yeshwanthpur, Bengaluru, India
| | - Wahaj Munir
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Lara Victoria Schmitt
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rohan Vyas
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jeuela Iris Ravindran
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mohamad Bashir
- Institue of Cardiac and Aortic Disorders, SRM Institutes for Medical Science (SIMS Hospitals), Chennai, India
| | | | - Bashi Velayudhan
- Institue of Cardiac and Aortic Disorders, SRM Institutes for Medical Science (SIMS Hospitals), Chennai, India
| | - Mohammed Idhrees
- Institue of Cardiac and Aortic Disorders, SRM Institutes for Medical Science (SIMS Hospitals), Chennai, India
| |
Collapse
|
102
|
Kimura Y, Tsukui D, Kono H. Uric Acid in Inflammation and the Pathogenesis of Atherosclerosis. Int J Mol Sci 2021; 22:ijms222212394. [PMID: 34830282 PMCID: PMC8624633 DOI: 10.3390/ijms222212394] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/06/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Hyperuricemia is a common metabolic syndrome. Elevated uric acid levels are risk factors for gout, hypertension, and chronic kidney diseases. Furthermore, various epidemiological studies have also demonstrated an association between cardiovascular risks and hyperuricemia. In hyperuricemia, reactive oxygen species (ROS) are produced simultaneously with the formation of uric acid by xanthine oxidases. Intracellular uric acid has also been reported to promote the production of ROS. The ROS and the intracellular uric acid itself regulate several intracellular signaling pathways, and alterations in these pathways may result in the development of atherosclerotic lesions. In this review, we describe the effect of uric acid on various molecular signals and the potential mechanisms of atherosclerosis development in hyperuricemia. Furthermore, we discuss the efficacy of treatments for hyperuricemia to protect against the development of atherosclerosis.
Collapse
Affiliation(s)
- Yoshitaka Kimura
- Department of Internal Medicine, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan; (Y.K.); (D.T.)
- Department of Microbiology and Immunology, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan
| | - Daisuke Tsukui
- Department of Internal Medicine, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan; (Y.K.); (D.T.)
| | - Hajime Kono
- Department of Internal Medicine, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan; (Y.K.); (D.T.)
- Correspondence: ; Tel.: +81-3-3964-1211
| |
Collapse
|
103
|
Calcium Dobesilate Modulates PKCδ-NADPH Oxidase- MAPK-NF-κB Signaling Pathway to Reduce CD14, TLR4, and MMP9 Expression during Monocyte-to-Macrophage Differentiation: Potential Therapeutic Implications for Atherosclerosis. Antioxidants (Basel) 2021; 10:antiox10111798. [PMID: 34829669 PMCID: PMC8615002 DOI: 10.3390/antiox10111798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Monocyte-to-macrophage differentiation results in the secretion of various inflammatory mediators and oxidative stress molecules necessary for atherosclerosis pathogenesis. Consequently, this differentiation represents a potential clinical target in atherosclerosis. Calcium dobesilate (CaD), an established vasoactive and angioprotective drug in experimental models of diabetic microvascular complications reduces oxidative stress and inhibits inflammation via diverse molecular targets; however, its effect on monocytes/macrophages is poorly understood. In this study, we investigated the anti-inflammatory mechanism of CaD during phorbol 12-myristate 13-acetate (PMA)-induced monocyte-to-macrophage differentiation in in vitro models of sepsis (LPS) and hyperglycemia, using THP-1 monocytic cell line. CaD significantly suppressed CD14, TLR4, and MMP9 expression and activity, lowering pro-inflammatory mediators, such as IL1β, TNFα, and MCP-1. The effects of CaD translated through to studies on primary human macrophages. CaD inhibited reactive oxygen species (ROS) generation, PKCδ, MAPK (ERK1/2 and p38) phosphorylation, NOX2/p47phox expression, and membrane translocation. We used hydrogen peroxide (H2O2) to mimic oxidative stress, demonstrating that CaD suppressed PKCδ activation via its ROS-scavenging properties. Taken together, we demonstrate for the first time that CaD suppresses CD14, TLR4, MMP9, and signature pro-inflammatory cytokines, in human macrophages, via the downregulation of PKCδ/NADPH oxidase/ROS/MAPK/NF-κB-dependent signaling pathways. Our data present novel mechanisms of how CaD alleviates metabolic and infectious inflammation.
Collapse
|
104
|
Liu C, Du H, Wang Y, Gong N, Qi W, Zhou X, Shi L. S100A11 regulates nasal epithelial cell remodeling and inflammation in CRSwNPs via the RAGE-mediated AMPK-STAT3 pathway. Mol Immunol 2021; 140:35-46. [PMID: 34653793 DOI: 10.1016/j.molimm.2021.09.014] [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: 05/26/2021] [Revised: 08/25/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022]
Abstract
Abnormal remodeling of the nasal mucosal epithelium and persistent chronic inflammation are important pathological features of chronic sinusitis with nasal polyps (CRSwNPs). In order to explore the molecular regulation mechanism of CRSwNPs, we performed iTRAQ protein profile analysis on 18 clinical samples collected (9 patients with nasal polyps and 9 healthy patients) and found that S100A11, a Ca2+-binding protein, was significantly higher in CRSwNPs. Subsequently, we demonstrated that S100A11 was mainly located in nasal mucosal epithelial cells and is up-regulated in human nasal epithelial stem/progenitor cells (hNESPCs) from CRSwNPs patients and CRSwNPs epithelial cell model established with S. aureus. To determine the functional role of S100A11 and the signal pathways in epithelial cells, we constructed S100A11 overexpression vector, small interfering RNA, recombinant protein-S100A11 (rh-S100A11) and RAGE inhibitor (sRAGE). Results showed that upregulation of S100A11 inhibited epithelial cell viability and promoted apoptosis and inflammation, in addition, S100A11 can regulate the signal homeostasis of AMPK-STAT3 via RAGE mediation in epithelial cells. Our findings suggest that S100A11 is involved in CRSwNPs epithelial tissue remodeling and inflammatory response regulation and may be a useful target for CRSwNPs therapy.
Collapse
Affiliation(s)
- Chengcheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250022, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Weiqi Road, Jinan, Shandong, China
| | - Hongjie Du
- Qilu Pharmaceutical Co.Ltd, 8888, Lvyou Rd Jinan, Shandong Province, China
| | - Yajie Wang
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Weiqi Road, Jinan, Shandong, China
| | - Ningyue Gong
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Weiqi Road, Jinan, Shandong, China
| | - Wenwen Qi
- Department of Otolaryngology, The Second Hospital of Shandong University, Shandong University, 274 Beiyuan Road, Jinan, Shandong, China
| | - Xiangmin Zhou
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250022, China
| | - Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250022, China.
| |
Collapse
|
105
|
Xiong W, Sun KY, Zhu Y, Zhang X, Zhou YH, Zou X. Metformin alleviates inflammation through suppressing FASN-dependent palmitoylation of Akt. Cell Death Dis 2021; 12:934. [PMID: 34642298 PMCID: PMC8511025 DOI: 10.1038/s41419-021-04235-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 12/18/2022]
Abstract
Metformin, traditionally regarded as a hypoglycemic drug, has been studied in other various fields including inflammation. The specific mechanism of metformin’s effect on immune cells remains unclear. Herein, it is verified that LPS-induced macrophages are characterized by enhanced endogenous fatty acid synthesis and the inhibition of fatty acid synthase (FASN) downregulates proinflammatory responses. We further show that metformin could suppress such elevation of FASN as well as proinflammatory activation in macrophages. In vivo, metformin treatment ameliorates dextran sulfate sodium (DSS)-induced colitis through impairing proinflammatory activation of colonic lamina propria mononuclear cells (LPMCs). The reduction of FASN by metformin hinders Akt palmitoylation, which further disturbs Akt membrane attachment and its phosphorylation. Metformin-mediated suppression of FASN/Akt pathway and its downstream MAPK signaling contributes to its anti-inflammatory role in macrophages. From the perspective of immunometabolism, our work points towards metformin utilization as an effective and potential intervention against macrophages-involved inflammatory diseases.
Collapse
Affiliation(s)
- Wenfang Xiong
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Kuo-Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Yan Zhu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Xiaoqi Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Yi-Hua Zhou
- Departments of Laboratory Medicine and Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
106
|
Kunath A, Unosson J, Friederich-Persson M, Bjarnegård N, Becirovic-Agic M, Björck M, Mani K, Wanhainen A, Wågsäter D. Inhibition of angiotensin-induced aortic aneurysm by metformin in apolipoprotein E-deficient mice. JVS Vasc Sci 2021; 2:33-42. [PMID: 34617056 PMCID: PMC8489247 DOI: 10.1016/j.jvssci.2020.11.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/30/2020] [Indexed: 10/26/2022] Open
Abstract
Objective Metformin is associated with a reduced incidence and growth of abdominal aortic aneurysms (AAAs). The aim of the present study was to investigate the inhibitory effects of metformin on AAA development and possible underlying mechanisms in experimentally induced AAAs in mice, along with the possible synergistic effects of metformin and imatinib. Methods Angiotensin II was used to induce AAAs in apolipoprotein E knockout (ApoE -/- ) mice for 28 days. The mice were treated with metformin (n = 11), metformin combined with imatinib (n = 7), or vehicle (n = 12), starting 3 days before angiotensin II infusion. Ultrasound examination was used to analyze aneurysm formation. Cholesterol and blood pressure levels were measured at the start and end of the study. Gene array and quantitative polymerase chain reaction were used to analyze the changes in gene expression in the aorta. Wire myography was used to study vascular function. Results Metformin (n = 11) suppressed the formation and progression of AAAs by 50% compared with the vehicle controls (n = 12), with no further effects from imatinib (n = 7). Metformin reduced total cholesterol and mRNA expression of SPP1 (encoding osteopontin), MMP12, and the glycoprotein genes Gpnmb and Clec7a. Furthermore, metformin inhibited blood pressure increases and reduced vascular contractions, as determined by wire myography, and restored the anticontractile function of perivascular adipose tissue. Conclusion Metformin inhibited aneurysm formation and progression and normalized vascular function in ApoE -/- mice with no additional effect of imatinib. This might be mediated by the protective effects on vascular endothelial function and perivascular adipose tissue via reduced expression of genes promoting inflammation, including SPP1, MMP12, Gpnmb, and Clec7a. Clinical relevance Retrospective studies of the effects of metformin in patients with aneurysm have so far only been performed of those with type 2 diabetes. The present study shows that metformin has effects on nondiabetic mice and revealed the mechanistic effects mediated by the drug that could also be important to study as outcomes in humans. Future clinical trials using metformin are warranted in patients without diabetes with abdominal aortic aneurysms.
Collapse
Affiliation(s)
- Anne Kunath
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Jon Unosson
- Section of Vascular Surgery, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Niclas Bjarnegård
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | | | - Martin Björck
- Section of Vascular Surgery, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Kevin Mani
- Section of Vascular Surgery, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Wanhainen
- Section of Vascular Surgery, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Dick Wågsäter
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
107
|
Heo E, Kim E, Jang EJ, Lee CH. The cumulative dose-dependent effects of metformin on the development of tuberculosis in patients newly diagnosed with type 2 diabetes mellitus. BMC Pulm Med 2021; 21:303. [PMID: 34563159 PMCID: PMC8464151 DOI: 10.1186/s12890-021-01667-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a well-known risk factor for tuberculosis (TB). Metformin, which is an essential anti-diabetic drug, has been shown to exhibit anti-TB effects in patients with DM. Its effect on preventing the development of TB among patients who are newly diagnosed with DM remains unclear. METHODS This was a retrospective cohort study using the claims database of the Korean Health Insurance Review and Assessment Service. The study population included patients who were newly diagnosed with type 2 DM and who were treated with anti-diabetic drugs between 1 January 2003 and 31 March 2011. A patient was defined as a metformin user if he/she had taken metformin for more than 28 days within 6 months since cohort entry, and as a metformin non-user if he/she had never been treated with metformin. The development of TB within 2 years after the index date was compared by Cox proportional hazard regression models between metformin users and 1:1 propensity score (PS)-matched non-users. RESULTS Among 76,973 patients who were newly diagnosed with type 2 DM, 13,396 were classified as metformin users, 52,736 were classified as metformin non-users, and 10,841 were excluded from the final analysis. PS-matched Cox proportional hazard regression models revealed that metformin use was not associated overall with the prevention of TB development (HR 1.17; 95% CI 0.75-1.83; P = 0.482). There was a trend, however, towards a reduction in the development of TB among patients taking a higher cumulative dose of metformin. Patients who were in the highest quartile (Q4) of cumulative metformin dose had only a 10% risk of developing TB compared to metformin non-users. In contrast, during the early phases of metformin treatment, patients in the second quartile (Q2) of cumulative metformin use had a higher risk of developing TB than patients in the first quartile (Q1). CONCLUSIONS Only the highest cumulative doses of metformin were protective against the development of TB among patients who were newly diagnosed with type 2 DM; lower cumulative doses of metformin did not appear to reduce the incidence of active TB infection.
Collapse
Affiliation(s)
- Eunyoung Heo
- Department of Internal Medicine, SNU-SMG Boramae Medical Center, Seoul, Republic of Korea
| | - Eunyoung Kim
- Department of Statistics, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Jin Jang
- Department of Information Statistics, Andong National University, Andong, Republic of Korea
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
108
|
Thanigaimani S, Singh TP, Unosson J, Phie J, Moxon J, Wanhainen A, Golledge J. Editor's Choice - Association Between Metformin Prescription and Abdominal Aortic Aneurysm Growth and Clinical Events: a Systematic Review and Meta-Analysis. Eur J Vasc Endovasc Surg 2021; 62:747-756. [PMID: 34556425 DOI: 10.1016/j.ejvs.2021.06.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/06/2021] [Accepted: 06/13/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE A meta-analysis of the association between metformin prescription and abdominal aortic aneurysm (AAA) growth and events (rupture or surgical repair) was performed. METHODS Open source databases were searched for observational studies reporting the association between metformin prescription and AAA growth or events. Meta-analyses were performed using random effects models. The risk of bias of included studies was assessed using a quality assessment tool developed in a previous systematic review. Sensitivity analyses restricted to people with diabetes, leave one out analyses, and an individual patient risk factor adjusted sub-analysis were performed. Funnel plots assessed reporting bias. RESULTS Eight studies comprising 153 553 patients were included, of whom 35 240 were and 118 313 were not prescribed metformin. Pooled weighted mean (± standard deviation) AAA growth was significantly reduced in patients prescribed metformin (0.9 ± 0.4 mm/year) compared with those not receiving the medication (1.8 ± 0.4 mm/year; weighted mean difference [WMD] 0.8 mm/year, 95% confidence interval [CI] 0.5 - 1.1; p < .001; I2 = 89%). Leave one out analysis suggested that the significance of findings did not change after removal of individual studies. A sub-analysis within people with diabetes suggested that metformin reduced AAA growth (WMD 0.7 mm/year, 95% CI 0.3 - 1.0). Metformin prescription was associated with a reduced risk of AAA events (risk ratio 0.6, 95% CI 0.4 - 0.9, p = .028). Three, four, and one studies had low, moderate, and high risk of bias, respectively. Individual patient data analysis suggested that metformin prescription slowed annual AAA growth by 0.5 mm/year (95% CI 0.2 - 0.7). The GRADE summary suggested that the certainty of evidence that metformin limited AAA growth and prevented AAA events was very low. CONCLUSION Observational studies suggest that metformin prescription is associated with a clinically important significant reduction in both growth and clinically relevant events in people with AAA. These findings support the need for randomised trials to examine the benefit of metformin.
Collapse
Affiliation(s)
- Shivshankar Thanigaimani
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Queensland, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Tejas P Singh
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Queensland, Australia; The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Queensland, Australia
| | - Jon Unosson
- Department of Surgical Sciences, Vascular Surgery, Uppsala University, Uppsala, Sweden
| | - James Phie
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Queensland, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Joseph Moxon
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Queensland, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Anders Wanhainen
- Department of Surgical Sciences, Vascular Surgery, Uppsala University, Uppsala, Sweden
| | - Jonathan Golledge
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Queensland, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia; The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Queensland, Australia.
| |
Collapse
|
109
|
Hu X, Luo H, Dou C, Chen X, Huang Y, Wang L, Xue S, Sun Z, Chen S, Xu Q, Geng T, Zhao X, Cui H. Metformin Triggers Apoptosis and Induction of the G0/G1 Switch 2 Gene in Macrophages. Genes (Basel) 2021; 12:genes12091437. [PMID: 34573418 PMCID: PMC8468785 DOI: 10.3390/genes12091437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin is a widely used antidiabetic drug for the treatment of type 2 diabetes and has been recently demonstrated to possess anti-inflammatory properties via AMPK-mediated modulation of M2 macrophage activation. However, the anti-inflammatory mechanisms of metformin on inflammatory macrophages are still not fully elucidated. In this study, we found that metformin induced apoptosis in macrophages. In particular, metformin induced apoptosis of M1 macrophages, based on M1 marker genes in apoptotic macrophages. Next, we comprehensively screened metformin-responsive genes in macrophages by RNA-seq and focused on the extrinsic apoptotic signaling pathway. The G0/G1 switch 2 gene (G0S2) was robustly up-regulated by metformin in macrophages. Overexpression of G0S2 significantly induced apoptosis of macrophages in a dose-dependent manner and blunted the function of the crucial anti-apoptotic gene Bcl-2, which was significantly reduced by metformin. These findings show that metformin promoted apoptosis of macrophages, especially M1 macrophages, via G0S2 induction and provides a novel anti-inflammatory mechanism of metformin through induction of macrophage apoptosis.
Collapse
Affiliation(s)
- Xuming Hu
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
- Department of Animal Science, McGill University, Montréal, QC H3A 0G4, Canada;
| | - Huan Luo
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Chunfeng Dou
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Xujing Chen
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Yi Huang
- Department of Pharmacy, Suzhou Vocational Health College, Suzhou 215009, China;
| | - Liping Wang
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Songlei Xue
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Zhen Sun
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Shihao Chen
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Qi Xu
- Department of Animal Science, McGill University, Montréal, QC H3A 0G4, Canada;
| | - Tuoyu Geng
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Xin Zhao
- Department of Animal Science, McGill University, Montréal, QC H3A 0G4, Canada;
- Correspondence: (X.Z.); (H.C.)
| | - Hengmi Cui
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention & Control of Important Animal Infectious Diseases & Zoonoses, Yangzhou 225009, China
- Correspondence: (X.Z.); (H.C.)
| |
Collapse
|
110
|
Borgoni S, Kudryashova KS, Burka K, de Magalhães JP. Targeting immune dysfunction in aging. Ageing Res Rev 2021; 70:101410. [PMID: 34280555 DOI: 10.1016/j.arr.2021.101410] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/28/2021] [Accepted: 07/14/2021] [Indexed: 12/23/2022]
Abstract
Human aging is a multifactorial phenomenon that affects numerous organ systems and cellular processes, with the immune system being one of the most dysregulated. Immunosenescence, the gradual deterioration of the immune system, and inflammaging, a chronic inflammatory state that persists in the elderly, are among the plethora of immune changes that occur during aging. Almost all populations of immune cells change with age in terms of numbers and/or activity. These alterations are in general highly detrimental, resulting in an increased susceptibility to infections, reduced healing abilities, and altered homeostasis that promote the emergence of age-associated diseases such as cancer, diabetes, and other diseases associated with inflammation. Thanks to recent developments, several strategies have been proposed to target central immunological processes or specific immune subpopulations affected by aging. These therapeutic approaches could soon be applied in the clinic to slow down or even reverse specific age-induced immune changes in order to rejuvenate the immune system and prevent or reduce the impact of various diseases. Due to its systemic nature and interconnection with all the other systems in the body, the immune system is an attractive target for aging intervention because relatively targeted modifications to a small set of cells have the potential to improve the health of multiple organ systems. Therefore, anti-aging immune targeting therapies could represent a potent approach for improving healthspan. Here, we review aging changes in the major components of the immune system, we summarize the current immune-targeting therapeutic approaches in the context of aging and discuss the future directions in the field of immune rejuvenation.
Collapse
|
111
|
Cheng D, Xu Q, Wang Y, Li G, Sun W, Ma D, Zhou S, Liu Y, Han L, Ni C. Metformin attenuates silica-induced pulmonary fibrosis via AMPK signaling. J Transl Med 2021; 19:349. [PMID: 34399790 PMCID: PMC8365894 DOI: 10.1186/s12967-021-03036-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Silicosis is one of the most common occupational pulmonary fibrosis caused by respirable silica-based particle exposure, with no ideal drugs at present. Metformin, a commonly used biguanide antidiabetic agent, could activate AMP-activated protein kinase (AMPK) to exert its pharmacological action. Therefore, we sought to investigate the role of metformin in silica-induced lung fibrosis. Methods The anti-fibrotic role of metformin was assessed in 50 mg/kg silica-induced lung fibrosis model. Silicon dioxide (SiO2)-stimulated lung epithelial cells/macrophages and transforming growth factor-beta 1 (TGF-β1)-induced differentiated lung fibroblasts were used for in vitro models. Results At the concentration of 300 mg/kg in the mouse model, metformin significantly reduced lung inflammation and fibrosis in SiO2-instilled mice at the early and late fibrotic stages. Besides, metformin (range 2–10 mM) reversed SiO2-induced cell toxicity, oxidative stress, and epithelial-mesenchymal transition process in epithelial cells (A549 and HBE), inhibited inflammation response in macrophages (THP-1), and alleviated TGF-β1-stimulated fibroblast activation in lung fibroblasts (MRC-5) via an AMPK-dependent pathway. Conclusions In this study, we identified that metformin might be a potential drug for silicosis treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03036-5.
Collapse
Affiliation(s)
- Demin Cheng
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qi Xu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yue Wang
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Guanru Li
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wenqing Sun
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Dongyu Ma
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Siyun Zhou
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Liu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Lei Han
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210028, China.
| | - Chunhui Ni
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
112
|
Savarese G, Butler J, Lund LH, Bhatt DL, Anker SD. CARDIOVASCULAR EFFECTS OF NON-INSULIN GLUCOSE-LOWERING AGENTS: A COMPREHENSIVE REVIEW OF TRIAL EVIDENCE AND POTENTIAL CARDIOPROTECTIVE MECHANISMS. Cardiovasc Res 2021; 118:2231-2252. [PMID: 34390570 DOI: 10.1093/cvr/cvab271] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/12/2021] [Indexed: 11/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is highly prevalent and associated with a 2-fold increased mortality, mostly explained by cardiovascular diseases. Trial evidence on older glucose-lowering agents such as metformin and sulfonylureas is limited in terms of cardiovascular efficacy. Since 2008, after rosiglitazone was observed to increase the risk of myocardial infarction and heart failure (HF), cardiovascular outcome trials (CVOT) have been required by regulators for licensing new glucose-lowering agents. In the following CVOTs, dipeptidyl peptidase 4 inhibitors (DPP4i) have been shown to be safe but not to improve morbidity/mortality, except for saxagliptin which increased the risk of HF. Several glucagon-like peptide-1 receptor agonists (GLP1-Ra) and sodium-glucose cotransporter-2 inhibitors (SGLT2i) have been demonstrated to reduce the risk of cardiovascular morbidity and mortality. SGLT2i have shown a class effect for the reduction in risk of HF events in patients with T2DM, leading to trials testing their efficacy/safety in HF regardless of T2DM. In the DAPA-HF and the EMPEROR-Reduced trials dapagliflozin and empagliflozin, respectively, improved cardiovascular mortality/morbidity in patients with HF and reduced ejection fraction (HFrEF), with and without T2DM. Therefore, these drugs are now key part of HFrEF pharmacotherapy. In the SOLOIST-WHF, sotagliflozin reduced cardiovascular mortality/morbidity in patients with T2DM and a recent acute episode of HF regardless of EF. The DELIVER and the EMPEROR-Preserved are testing dapagliflozin and empagliflozin, respectively, in patients with HF with mildly reduced and preserved EF. A strong renal protective role of SGLT2i has also emerged in trials enrolling patients with and without T2DM.
Collapse
Affiliation(s)
- Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden. Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Javed Butler
- University of Mississippi School of Medicine, Jackson, MI, USA
| | - Lars H Lund
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden. Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart and Vascular Center and Harvard Medical School, Boston, MA, USA
| | - Stefan D Anker
- Department of Cardiology (CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT), and German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin Berlin, Germany
| |
Collapse
|
113
|
Sun Z, Li J, Luo G, Liu W, He Y, Wang F, Qian Y, Fan C. Pharmacological activation of SIRT1 by metformin prevented trauma-induced heterotopic ossification through inhibiting macrophage mediated inflammation. Eur J Pharmacol 2021; 909:174386. [PMID: 34332919 DOI: 10.1016/j.ejphar.2021.174386] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Trauma-induced heterotopic ossification (HO) is the aberrant extra-skeletal bone formation that severely incapacitates patient's daily life. Inflammation is the first stage of this progression, becoming an appealing target of early therapeutic intervention. Metformin, a widely used antidiabetic drug, also poses the therapeutic potential to modulate various inflammatory-related diseases. Therefore, this study aimed to investigate the preventive effect of metformin on trauma-induced HO progression, and unveil the underlying molecular mechanisms. A murine burn/tenotomy model was established to mimic trauma-induced HO in vivo. The anti-inflammation and anti-ossification effects of metformin were evaluated by histological staining and micro-CT. The inhibitory effects of metformin on macrophages activation in vitro were examined by ELISA and qRT-PCR. The underlying molecular mechanisms were further explored by immunofluorescence staining and western-blotting in vivo. Increased macrophages infiltration and inflammatory responses were found at early stage during HO progression. However, metformin dose-dependently attenuated the macrophage-mediated inflammatory responses both in vivo and vitro, which might account for the inhibitory effect of metformin on chondrogenesis and HO formation after trauma. Furthermore, elevated SIRT1 expression and decreased NF-κB p65 acetylation were found in the beneficial effects of metformin. Moreover, similar preventive effects were also found in SRT1720 HCI, a specific SIRT1 activator, while were remarkably reversed after the administration of EX527 (a specific SIRT1 inhibitor) with metformin. Taken together, our results provide a novel evidence that metformin can effectively attenuate trauma-induced HO by mitigating macrophage inflammatory responses through inhibiting NF-κB signaling via SIRT1-dependent mechanisms, which favors future therapeutic investigations for trauma-related disease.
Collapse
Affiliation(s)
- Ziyang Sun
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Juehong Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Gang Luo
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Weixuan Liu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Yunwei He
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Feiyan Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China.
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China.
| |
Collapse
|
114
|
Palmer TM, Salt IP. Nutrient regulation of inflammatory signalling in obesity and vascular disease. Clin Sci (Lond) 2021; 135:1563-1590. [PMID: 34231841 DOI: 10.1042/cs20190768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/10/2021] [Accepted: 06/28/2021] [Indexed: 11/17/2022]
Abstract
Despite obesity and diabetes markedly increasing the risk of developing cardiovascular diseases, the molecular and cellular mechanisms that underlie this association remain poorly characterised. In the last 20 years it has become apparent that chronic, low-grade inflammation in obese adipose tissue may contribute to the risk of developing insulin resistance and type 2 diabetes. Furthermore, increased vascular pro-inflammatory signalling is a key event in the development of cardiovascular diseases. Overnutrition exacerbates pro-inflammatory signalling in vascular and adipose tissues, with several mechanisms proposed to mediate this. In this article, we review the molecular and cellular mechanisms by which nutrients are proposed to regulate pro-inflammatory signalling in adipose and vascular tissues. In addition, we examine the potential therapeutic opportunities that these mechanisms provide for suppression of inappropriate inflammation in obesity and vascular disease.
Collapse
Affiliation(s)
- Timothy M Palmer
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, United Kingdom
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
115
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 881] [Impact Index Per Article: 293.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
116
|
Rahmatpour Rokni G, Shiran M, Abounoori M, Houshmand G, Babakhanian M, Godazandeh G, Bayat S, Pazyar N, Abedi M, Khorshidi F, Yari F, Ghafouri Z, Patil A, Goldust M, Mirmohammadi Langari L. Effects of metformin on autoimmune immunoglobins and interferon-γ in patients with early diagnosed pemphigus vulgaris: a prospective clinical trial. Clin Exp Dermatol 2021; 47:110-113. [PMID: 34236726 DOI: 10.1111/ced.14832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 11/27/2022]
Abstract
The management of pemphigus vulgaris (PV) is challenging. This study aimed to evaluate the immunomodulating effects of metformin on PV. The study was conducted in two phases: in the first phase, patients received routine first-line treatment (prednisolone plus azathioprine) for 2 months, then in the second phase, metformin was added to this regimen for another 2 months. After addition of metformin to the first-line medications, significant reductions were seen in serum IgG1 (reduced from 534.92 ± 134.83 mg/dL to 481.58 ± 130.46 mg/dL, P < 0.001), IgG4 (51.83 ± 27.26 mg/dL to 44.50 ± 26.05 mg/dL, P < 0.001) and interferon-γ (277.99 ± 108.71 pg/mL to 45.05 ± 17.080 pg/mL, P = 0.03) concentrations. The suppressant effect of metformin was greatest on IgG4 (coefficient of variation 1.28), the dominant subclass of IgG involved in PV. Metformin could have immunomodulating effects on PV with controlling effects on steroid complications.
Collapse
Affiliation(s)
- G Rahmatpour Rokni
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - M Shiran
- Department of Pharmacology, Immuno-Genetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - M Abounoori
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - G Houshmand
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - M Babakhanian
- Social Determinants of Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - G Godazandeh
- Department of Thoracic Surgery, Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - S Bayat
- Student Research Committee, Department of Internal Medicine, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - N Pazyar
- Department of Dermatology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - M Abedi
- Faculty of Medicine, Azad University of Mazandaran, Sari, Iran
| | - F Khorshidi
- Faculty of Medicine, Azad University of Mazandaran, Sari, Iran
| | - F Yari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Z Ghafouri
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - A Patil
- Department of Pharmacology, Dr DY Patil Medical College, Navi Mumbai, India
| | - M Goldust
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | - L Mirmohammadi Langari
- Microbial Resistance Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
117
|
Elzahhar PA, Alaaeddine RA, Nassra R, Ismail A, Labib HF, Temraz MG, Belal ASF, El-Yazbi AF. Challenging inflammatory process at molecular, cellular and in vivo levels via some new pyrazolyl thiazolones. J Enzyme Inhib Med Chem 2021; 36:669-684. [PMID: 33618602 PMCID: PMC7901699 DOI: 10.1080/14756366.2021.1887169] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The work reported herein describes the synthesis of a new series of anti-inflammatory pyrazolyl thiazolones. In addition to COX-2/15-LOX inhibition, these hybrids exerted their anti-inflammatory actions through novel mechanisms. The most active compounds possessed COX-2 inhibitory activities comparable to celecoxib (IC50 values of 0.09-0.14 µM) with significant 15-LOX inhibitory activities (IC50s 1.96 to 3.52 µM). Upon investigation of their in vivo anti-inflammatory activities and ulcerogenic profiles, these compounds showed activity patterns equivalent or more superior to diclofenac and/or celecoxib. Intriguingly, the most active compounds were more effective than diclofenac in suppressing monocyte-to-macrophage differentiation and inflammatory cytokine production by activated macrophages, as well as their ability to induce macrophage apoptosis. The latter finding potentially adds a new dimension to the previously reported anti-inflammatory mechanisms of similar compounds. These compounds were effectively docked into COX-2 and 15-LOX active sites. Also, in silico predictions confirmed the appropriateness of these compounds as drug-like candidates.
Collapse
Affiliation(s)
- Perihan A Elzahhar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rana A Alaaeddine
- Department of Pharmacology and Toxicology, Faculty of Medicine and Medical Centre, American University of Beirut, Beirut, Lebanon
| | - Rasha Nassra
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Azza Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Hala F Labib
- Department of Pharmaceutical Chemistry, College of Pharmacy, Arab Academy of Science Technology and Maritime Transport, Alexandria, Egypt
| | | | - Ahmed S F Belal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine and Medical Centre, American University of Beirut, Beirut, Lebanon.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, E gypt
| |
Collapse
|
118
|
Kang K, Xia A, Meng F, Chunyu J, Sun X, Ren G, Yu D, Jiang X, Tang L, Xiao W, Li D. FGF21 alleviates chronic inflammatory injury in the aging process through modulating polarization of macrophages. Int Immunopharmacol 2021; 96:107634. [PMID: 33872851 DOI: 10.1016/j.intimp.2021.107634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 01/14/2023]
Abstract
Previous studies reported that FGF21 prolongs life span and delays the body senescence, but the mechanism is not clear. The present study was designed to investigate the effects of FGF21 on hepatic senescence in aging mice and further research the mechanism. The 14-month-old male mice were administered with PBS, FGF21 or metformin once daily for 6 months. Results showed that FGF21 alleviated liver injury and inhibited accumulation of senescence markers SASP, P53 and P16 in the livers of aging mice. Subsequently we found that the aging mice treated by FGF21 showed transition of type 1 macrophages (M1) to type 2 macrophages (M2) in the livers. Next, we used THP-1 macrophages triggered by LPS to study effects of FGF21 on macrophages. Macrophages triggered by LPS exhibited features of M1, but the addition of FGF21 decreased the expression of M1 markers, and promoted the macrophages to exhibit features of M2. Results showed that the effects of FGF21 on macrophages were associated with the AMPK pathway. After adding AMPK inhibitor, the effects of FGF21 were inhibited, which was associated with the NF-κB signaling pathway. Finally, co-culturing differentiated macrophages and hepatocytes, we found that the large amount of pro-inflammatory factors such as IL-6 promoted hepatocyte senescence, which exhibited enhanced P53, P16 and β-galactosidase. This was contrary to hepatocytes co-cultured with macrophages treated by FGF21. These results indicate that FGF21 alleviates hepatic senescence injury by modulating the polarization of macrophages through the AMPK /NF-κB signaling pathway.
Collapse
Affiliation(s)
- Kai Kang
- Northeast Agricultural University, Harbin, China.
| | - Anran Xia
- Northeast Agricultural University, Harbin, China.
| | - Fanrui Meng
- Northeast Agricultural University, Harbin, China.
| | - Jian Chunyu
- Northeast Agricultural University, Harbin, China.
| | - Xu Sun
- Northeast Agricultural University, Harbin, China.
| | - Guiping Ren
- Northeast Agricultural University, Harbin, China.
| | - Dan Yu
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical CO. LTD, Lianyungang, Jiangsu, China.
| | | | - Lei Tang
- Harbin Weike Biotechnology CO. LTD, Harbin, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical CO. LTD, Lianyungang, Jiangsu, China.
| | - Deshan Li
- Northeast Agricultural University, Harbin, China.
| |
Collapse
|
119
|
Kaur N, Guan Y, Raja R, Ruiz-Velasco A, Liu W. Mechanisms and Therapeutic Prospects of Diabetic Cardiomyopathy Through the Inflammatory Response. Front Physiol 2021; 12:694864. [PMID: 34234695 PMCID: PMC8257042 DOI: 10.3389/fphys.2021.694864] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
The incidence of heart failure (HF) continues to increase rapidly in patients with diabetes. It is marked by myocardial remodeling, including fibrosis, hypertrophy, and cell death, leading to diastolic dysfunction with or without systolic dysfunction. Diabetic cardiomyopathy (DCM) is a distinct myocardial disease in the absence of coronary artery disease. DCM is partially induced by chronic systemic inflammation, underpinned by a hostile environment due to hyperglycemia, hyperlipidemia, hyperinsulinemia, and insulin resistance. The detrimental role of leukocytes, cytokines, and chemokines is evident in the diabetic heart, yet the precise role of inflammation as a cause or consequence of DCM remains incompletely understood. Here, we provide a concise review of the inflammatory signaling mechanisms contributing to the clinical complications of diabetes-associated HF. Overall, the impact of inflammation on the onset and development of DCM suggests the potential benefits of targeting inflammatory cascades to prevent DCM. This review is tailored to outline the known effects of the current anti-diabetic drugs, anti-inflammatory therapies, and natural compounds on inflammation, which mitigate HF progression in diabetic populations.
Collapse
Affiliation(s)
| | | | | | | | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
120
|
Singh A, Sen P. Lipid droplet: A functionally active organelle in monocyte to macrophage differentiation and its inflammatory properties. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158981. [PMID: 34119681 DOI: 10.1016/j.bbalip.2021.158981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/06/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022]
Abstract
Lipid droplets (LDs) perform several important functions like inflammatory responses, membrane trafficking, acts as secondary messengers, etc. rather than simply working as an energy reservoir. LDs have been implicated as a controlling factor in the progression of atherosclerosis followed by foam cell formation that derives from macrophages during the differentiation process. However, the role of LDs in monocyte differentiation or its further immunological function is still an area that mandates in-depth investigation. We report that LD dynamics is important for differentiation of monocytes and is absolutely required for sustained and prolonged functional activity of differentiated macrophages. In THP-1 cell line model system, we elucidated that increase in total LD content in monocyte by external lipid supplements, can induce monocyte differentiation independent of classical stimuli, PMA. Differential expression of PLIN2 and ATGL during the event, together with abrogation of de novo lipogenesis further confirmed the fact. Besides, an increase in LD content by free fatty acid supplement was able to exert a synergistic effect with PMA on differentiation and phagocytic activity compared to when they are used alone. Additionally, we have shown Rab5a to play a vital role in LDs biosynthesis/maturation in monocytes and thereby directly affecting differentiation of monocytes into macrophages via AKT pathway. Thus our study reveals the multi-faceted function of LDs during the process of monocyte to macrophage differentiation and thereby helping to maintain the functional activity.
Collapse
Affiliation(s)
- Arpana Singh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Prosenjit Sen
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India.
| |
Collapse
|
121
|
Feng X, Chen W, Ni X, Little PJ, Xu S, Tang L, Weng J. Metformin, Macrophage Dysfunction and Atherosclerosis. Front Immunol 2021; 12:682853. [PMID: 34163481 PMCID: PMC8215340 DOI: 10.3389/fimmu.2021.682853] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Metformin is one of the most widely prescribed hypoglycemic drugs and has the potential to treat many diseases. More and more evidence shows that metformin can regulate the function of macrophages in atherosclerosis, including reducing the differentiation of monocytes and inhibiting the inflammation, oxidative stress, polarization, foam cell formation and apoptosis of macrophages. The mechanisms by which metformin regulates the function of macrophages include AMPK, AMPK independent targets, NF-κB, ABCG5/8, Sirt1, FOXO1/FABP4 and HMGB1. On the basis of summarizing these studies, we further discussed the future research directions of metformin: single-cell RNA sequencing, neutrophil extracellular traps (NETs), epigenetic modification, and metformin-based combination drugs. In short, macrophages play an important role in a variety of diseases, and improving macrophage dysfunction may be an important mechanism for metformin to expand its pleiotropic pharmacological profile. In addition, the combination of metformin with other drugs that improve the function of macrophages (such as SGLT2 inhibitors, statins and IL-1β inhibitors/monoclonal antibodies) may further enhance the pleiotropic therapeutic potential of metformin in conditions such as atherosclerosis, obesity, cancer, dementia and aging.
Collapse
Affiliation(s)
- Xiaojun Feng
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Wenxu Chen
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Xiayun Ni
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Peter J. Little
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| | - Liqin Tang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| |
Collapse
|
122
|
Timmons JG, Greenlaw N, Boyle JG, Chaturvedi N, Ford I, Brouwers MCGJ, Tillin T, Hramiak I, Hughes AD, Jenkins AJ, Klein BEK, Klein R, Ooi TC, Rossing P, Stehouwer CDA, Sattar N, Colhoun HM, Petrie JR. Metformin and carotid intima-media thickness in never-smokers with type 1 diabetes: The REMOVAL trial. Diabetes Obes Metab 2021; 23:1371-1378. [PMID: 33591613 DOI: 10.1111/dom.14350] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 02/05/2023]
Abstract
AIM To determine whether metformin's effects on carotid artery intima-media thickness (cIMT) in type 1 diabetes differ according to smoking status. METHODS Regression model effect estimates for the effect of metformin versus placebo (double-blind) on carotid IMT were calculated as a subgroup analysis of the REMOVAL trial. RESULTS In 428 randomized participants (227 never-smokers, 201 ever-smokers), averaged mean carotid IMT progression (per year) was reduced by metformin versus placebo in never-smokers (-0.012 mm, 95% CI -0.021 to -0.002; p = .0137) but not in ever-smokers (0.003 mm, 95% CI -0.008 to 0.014; p = .5767); and similarly in non-current smokers (-0.008 mm, 95% CI -0.015 to -0.00001; p = .0497) but not in current smokers (0.013 mm, 95% CI -0.007 to 0.032; p = .1887). Three-way interaction terms (treatment*time*smoking status) were significant for never versus ever smoking (p = .0373, prespecified) and non-current versus current smoking (p = .0496, exploratory). Averaged maximal carotid IMT progression (per year) was reduced by metformin versus placebo in never-smokers (-0.020 mm, 95% CI -0.034 to -0.006; p = .0067) but not in ever-smokers (-0.006 mm, 95% CI -0.020 to 0.008; p = .4067), although this analysis was not supported by a significant three-way interaction term. CONCLUSIONS This subgroup analysis of the REMOVAL trial provides additional support for a potentially wider role of adjunct metformin therapy in cardiovascular risk management in type 1 diabetes, particularly for individuals who have never smoked cigarettes.
Collapse
Affiliation(s)
- Joseph G Timmons
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nicola Greenlaw
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - James G Boyle
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nish Chaturvedi
- Institute of Cardiovascular Science, University College London, London, UK
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Martijn C G J Brouwers
- Department of Internal Medicine and Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Therese Tillin
- Institute of Cardiovascular Science, University College London, London, UK
| | | | - Alun D Hughes
- Institute of Cardiovascular Science, University College London, London, UK
| | - Alicia J Jenkins
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Barbara E K Klein
- University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ron Klein
- University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Teik C Ooi
- Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Peter Rossing
- Steno Diabetes Center Copenhagen and the University of Copenhagen, Copenhagen, Denmark
| | - Coen D A Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; and, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Helen M Colhoun
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - John R Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
123
|
Baradaran Z, Vakilian A, Zare M, Hashemzehi M, Hosseini M, Dinpanah H, Beheshti F. Metformin improved memory impairment caused by chronic ethanol consumption during adolescent to adult period of rats: Role of oxidative stress and neuroinflammation. Behav Brain Res 2021; 411:113399. [PMID: 34087254 DOI: 10.1016/j.bbr.2021.113399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/22/2021] [Accepted: 05/30/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Adolescence is a crucial time for brain maturation. We investigated the protective effects of metformin (Met) on behavioral changes, oxidative stress, tumor necrosis factor alpha (TNF-α) and nitrite in adulthood induced by ethanol (Eth) consumption during adolescent to adult period of rats. MATERIALS AND METHODS The adolescence male rats (21 days old) were treated as: 1) Control, 2) Eth (Eth in drinking water (20 %)), 3-5) Eth-Met50, 100 and 150 mg/kg (Eth in drinking water and Met (50, 100, or 150 mg/kg). After 5 weeks treatment, Morris water maze (MMW) and passive avoidance (PA) tests were done. RESULTS The latency in the MWM test was higher and the latency to enter the dark chamber in the PA test was lower in the Eth group than in control. In Eth-Met100 and 150 groups, they were less than the Eth group. Malondialdehyde (MDA) and nitrite concentration in the hippocampus and cortex of the Eth group were higher than the control group. The thiol content and catalase and superoxide dismutase (SOD) activities in hippocampal and cortical tissues of the Eth group reduced compared to the control group. TNF-α was higher in hippocampal tissues of Eth group animals. Met reversed all of these effects. CONCLUSION Our findings showed that the protective effects of Met against chronic Eth consumption induced learning and memory impairment were accompanied by decreasing of TNF-a, nitrite and oxidative stress in adolescent rats.
Collapse
Affiliation(s)
- Zahra Baradaran
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Arefeh Vakilian
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mostafa Zare
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Milad Hashemzehi
- Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Dinpanah
- Department of Emergency Medicine, 9-Day Hospital, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
| |
Collapse
|
124
|
Justice JN, Gubbi S, Kulkarni AS, Bartley JM, Kuchel GA, Barzilai N. A geroscience perspective on immune resilience and infectious diseases: a potential case for metformin. GeroScience 2021; 43:1093-1112. [PMID: 32902818 PMCID: PMC7479299 DOI: 10.1007/s11357-020-00261-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/27/2020] [Indexed: 12/18/2022] Open
Abstract
We are in the midst of the global pandemic. Though acute respiratory coronavirus (SARS-COV2) that leads to COVID-19 infects people of all ages, severe symptoms and mortality occur disproportionately in older adults. Geroscience interventions that target biological aging could decrease risk across multiple age-related diseases and improve outcomes in response to infectious disease. This offers hope for a new host-directed therapeutic approach that could (i) improve outcomes following exposure or shorten treatment regimens; (ii) reduce the chronic pathology associated with the infectious disease and subsequent comorbidity, frailty, and disability; and (iii) promote development of immunological memory that protects against relapse or improves response to vaccination. We review the possibility of this approach by examining available evidence in metformin: a generic drug with a proven safety record that will be used in a large-scale multicenter clinical trial. Though rigorous translational research and clinical trials are needed to test this empirically, metformin may improve host immune defenses and confer protection against long-term health consequences of infectious disease, age-related chronic diseases, and geriatric syndromes.
Collapse
Affiliation(s)
- Jamie N Justice
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| | - Sriram Gubbi
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Ameya S Kulkarni
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jenna M Bartley
- Center on Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - George A Kuchel
- Center on Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| |
Collapse
|
125
|
Metformin Actions on the Liver: Protection Mechanisms Emerging in Hepatocytes and Immune Cells against NASH-Related HCC. Int J Mol Sci 2021; 22:ijms22095016. [PMID: 34065108 PMCID: PMC8126028 DOI: 10.3390/ijms22095016] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is strongly linked to the global epidemic of obesity and type 2 diabetes mellitus (T2DM). Notably, NAFLD can progress from the mildest form of simple steatosis to nonalcoholic steatohepatitis (NASH) that increases the risk for hepatocellular carcinoma (HCC), which is a malignancy with a dismal prognosis and rising incidence in the United States and other developed counties, possibly due to the epidemic of NAFLD. Metformin, the first-line drug for T2DM, has been suggested to reduce risks for several types of cancers including HCC and protect against NASH-related HCC, as revealed by epidemical studies on humans and preclinical studies on animal models. This review focuses on the pathogenesis of NASH-related HCC and the mechanisms by which metformin inhibits the initiation and progression of NASH-related HCC. Since the functional role of immune cells in liver homeostasis and pathogenesis is increasingly appreciated in developing anti-cancer therapies on liver malignancies, we discuss both the traditional targets of metformin in hepatocytes and the recently defined effects of metformin on immune cells.
Collapse
|
126
|
Lin Y, Huang M, Wang S, You X, Zhang L, Chen Y. PAQR11 modulates monocyte-to-macrophage differentiation and pathogenesis of rheumatoid arthritis. Immunology 2021; 163:60-73. [PMID: 33421113 PMCID: PMC8044334 DOI: 10.1111/imm.13303] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/15/2020] [Accepted: 12/19/2020] [Indexed: 12/23/2022] Open
Abstract
During inflammation or tissue injury, pro-inflammatory mediators attract migratory monocytes to inflammatory sites and monocyte-to-macrophage differentiation occurs to activate macrophages. We report here that PAQR11, a member of the progesterone and AdipoQ receptor family, regulates monocyte-to-macrophage differentiation in vitro and in vivo. Paqr11 gene was highly induced during monocyte-to-macrophage differentiation. Knockdown or deletion of Paqr11 inhibited monocyte differentiation but had little effect on macrophage polarization. Mechanistically, PAQR11 promoted cell survival as apoptosis was increased by Paqr11 knockdown or deletion. Activation of the MAPK signalling pathway was involved in the regulatory role of PAQR11 on monocyte differentiation and cell survival. C/EBPβ regulated the expression of Paqr11 at the transcriptional level. In mice, deletion of Paqr11 gene alleviated progression of collagen-induced rheumatoid arthritis. Thus, these results provide strong evidence that PAQR11 has a function in monocyte-to-macrophage differentiation and such function is related to autoimmune disease in vivo.
Collapse
Affiliation(s)
- Yijun Lin
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Meiqin Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Shuo Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Xue You
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Lingling Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| |
Collapse
|
127
|
Seneviratne A, Cave L, Hyde G, Moestrup SK, Carling D, Mason JC, Haskard DO, Boyle JJ. Metformin directly suppresses atherosclerosis in normoglycaemic mice via haematopoietic adenosine monophosphate-activated protein kinase. Cardiovasc Res 2021; 117:1295-1308. [PMID: 32667970 PMCID: PMC8064441 DOI: 10.1093/cvr/cvaa171] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 06/03/2018] [Accepted: 06/22/2020] [Indexed: 12/31/2022] Open
Abstract
AIMS Atherosclerotic vascular disease has an inflammatory pathogenesis. Heme from intraplaque haemorrhage may drive a protective and pro-resolving macrophage M2-like phenotype, Mhem, via AMPK and activating transcription factor 1 (ATF1). The antidiabetic drug metformin may also activate AMPK-dependent signalling. Hypothesis: Metformin systematically induces atheroprotective genes in macrophages via AMPK and ATF1, thereby suppresses atherogenesis. METHODS AND RESULTS Normoglycaemic Ldlr-/- hyperlipidaemic mice were treated with oral metformin, which profoundly suppressed atherosclerotic lesion development (P < 5 × 10-11). Bone marrow transplantation from AMPK-deficient mice demonstrated that metformin-related atheroprotection required haematopoietic AMPK [analysis of variance (ANOVA), P < 0.03]. Metformin at a clinically relevant concentration (10 μM) evoked AMPK-dependent and ATF1-dependent increases in Hmox1, Nr1h2 (Lxrb), Abca1, Apoe, Igf1, and Pdgf, increases in several M2-markers and decreases in Nos2, in murine bone marrow macrophages. Similar effects were seen in human blood-derived macrophages, in which metformin-induced protective genes and M2-like genes, suppressible by si-ATF1-mediated knockdown. Microarray analysis comparing metformin with heme in human macrophages indicated that the transcriptomic effects of metformin were related to those of heme, but not identical. Metformin-induced lesional macrophage expression of p-AMPK, p-ATF1, and downstream M2-like protective effects. CONCLUSION Metformin activates a conserved AMPK-ATF1-M2-like pathway in mouse and human macrophages, and results in highly suppressed atherogenesis in hyperlipidaemic mice via haematopoietic AMPK.
Collapse
Affiliation(s)
| | - Luke Cave
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Gareth Hyde
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - David Carling
- MRC London Institute of Medical Sciences, Imperial College London, UK
| | - Justin C Mason
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Dorian O Haskard
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
128
|
Adly Sadik N, Ahmed Rashed L, Ahmed Abd-El Mawla M. Circulating miR-155 and JAK2/STAT3 Axis in Acute Ischemic Stroke Patients and Its Relation to Post-Ischemic Inflammation and Associated Ischemic Stroke Risk Factors. Int J Gen Med 2021; 14:1469-1484. [PMID: 33911894 PMCID: PMC8071708 DOI: 10.2147/ijgm.s295939] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Background “Micro RNAs and their target genes recently have been identified to play a crucial role in the molecular pathogenesis of post-stroke ischemic cellular injury, which elucidates their new role in ischemic stroke diagnosis and therapy”. Thus, we evaluated the relative serum expression of miR-155, an inflammatory micro RNA, and the mRNAs (JAK2/STAT3) in acute ischemic stroke patients and its associations with the inflammatory cytokine TNF-α and different stroke risk factors. Subjects and Methods The relative expression of serum miR-155 and mRNAs (JAK2/STAT3) was assessed using RT-PCR, serum TNF-α was measured using ELIZA in 46 acute ischemic stroke patients and 50 control subjects. Receiver operating characteristic (ROC) curve was constructed to assess the specificity and sensitivity of circulating miR-155, JAK2/STAT3 as biomarkers for acute ischemic stroke. Results Circulating miR-155, JAK2/STAT3 were significantly up-regulated among stroke patients (8.5, 2.9, 4.2 fold respectively, P<0.001) with significant increase in TNF-α (263.8 ± 10.7 pg/mL, P <0.001). MiR-155, JAK2/STAT3 were positively correlated with TNF-α. MiR-155, JAK2/STAT3 were significantly increased in stroke patients and associated with risk factors such as hypertension, carotid atherosclerosis, and atrial fibrillation. Our study revealed that miR-155 has diagnostic accuracy for acute ischemic stroke where AUC=0.9, (P<0.001). Conclusion The elevated expressions of circulating miR-155, JAK2/STAT3, and TNF-α in acute ischemic stroke patients could trigger post-stroke cellular inflammation. MiR-155 could be used as potential inflammatory biomarker for acute ischemic stroke. However, further clinical studies are still needed to determine the exact role of miRNAs and different signal transduction expressions in the stage of acute ischemic stroke.
Collapse
Affiliation(s)
- Noha Adly Sadik
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
129
|
Xu J, Kitada M, Ogura Y, Koya D. Relationship Between Autophagy and Metabolic Syndrome Characteristics in the Pathogenesis of Atherosclerosis. Front Cell Dev Biol 2021; 9:641852. [PMID: 33937238 PMCID: PMC8083902 DOI: 10.3389/fcell.2021.641852] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/19/2021] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is the main cause of mortality in metabolic-related diseases, including cardiovascular disease and type 2 diabetes (T2DM). Atherosclerosis is characterized by lipid accumulation and increased inflammatory cytokines in the vascular wall, endothelial cell and vascular smooth muscle cell dysfunction and foam cell formation initiated by monocytes/macrophages. The characteristics of metabolic syndrome (MetS), including obesity, glucose intolerance, dyslipidemia and hypertension, may activate multiple mechanisms, such as insulin resistance, oxidative stress and inflammatory pathways, thereby contributing to increased risks of developing atherosclerosis and T2DM. Autophagy is a lysosomal degradation process that plays an important role in maintaining cellular metabolic homeostasis. Increasing evidence indicates that impaired autophagy induced by MetS is related to oxidative stress, inflammation, and foam cell formation, further promoting atherosclerosis. Basal and mild adaptive autophagy protect against the progression of atherosclerotic plaques, while excessive autophagy activation leads to cell death, plaque instability or even plaque rupture. Therefore, autophagic homeostasis is essential for the development and outcome of atherosclerosis. Here, we discuss the potential role of autophagy and metabolic syndrome in the pathophysiologic mechanisms of atherosclerosis and potential therapeutic drugs that target these molecular mechanisms.
Collapse
Affiliation(s)
- Jing Xu
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Department of Endocrinology and Metabolism, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Yoshio Ogura
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
130
|
Zhang Q, Yang M, Xiao Y, Han Y, Yang S, Sun L. Towards Better Drug Repositioning: Targeted Immunoinflammatory Therapy for Diabetic Nephropathy. Curr Med Chem 2021; 28:1003-1024. [PMID: 31701843 DOI: 10.2174/0929867326666191108160643] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 11/22/2022]
Abstract
Diabetic nephropathy (DN) is one of the most common and important microvascular complications of diabetes mellitus (DM). The main clinical features of DN are proteinuria and a progressive decline in renal function, which are associated with structural and functional changes in the kidney. The pathogenesis of DN is multifactorial, including genetic, metabolic, and haemodynamic factors, which can trigger a sequence of events. Controlling metabolic risks such as hyperglycaemia, hypertension, and dyslipidaemia is not enough to slow the progression of DN. Recent studies emphasized immunoinflammation as a critical pathogenic factor in the progression of DN. Therefore, targeting inflammation is considered a potential and novel treatment strategy for DN. In this review, we will briefly introduce the inflammatory process of DN and discuss the anti-inflammatory effects of antidiabetic drugs when treating DN.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shikun Yang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
131
|
Turowicz A, Kobecki J, Laskowska A, Wojciechowski J, Świątkowski F, Chabowski M. Association of Metformin and Abdominal Aortic Aneurysm Repair Outcomes. Ann Vasc Surg 2021; 75:390-396. [PMID: 33826959 DOI: 10.1016/j.avsg.2021.02.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/14/2021] [Accepted: 02/22/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Metformin is a commonly used drug in diabetes mellitus treatment. Recently it has been suggested that the use of metformin on diabetes mellitus patients may lower the prevalence and slow the progression of AAA (abdominal aortic aneurysm) as well as the risk of rupture related mortality. The aim of this studywas to investigate the impact of metformin treatment on the risk of AAA repair related mortality and surgical complications. METHODS In this retrospective study, the clinical data of 306 patients, including 77 patients with diabetes mellitus, who underwent abdominal aortic aneurysm repair has been analyzed. Treatment outcomes have been investigated. The diabetes and metformin prescription status has been obtained from the medical history. Patients were divided into three groups: diabetes-free individuals, diabetics treated with metformin and diabetics treated with other glucose lowering drugs. The association between metformin treatment and AAA diameter, surgical complications and mortality were assessed using chi-square independence test and odds ratio analysis. In order to assess which factors are influencing AAA repair related complications and mortality a multi-variables analysis has been performed. RESULTS A significant protective effect of metformin treatment towards AAA repair related mortality (P = 0.019) and complications (P = 0.032) among patients suffering from diabetes mellitus was revealed. These findings were statistically insignificant when considering all groups of patients (diabetes-free individuals, diabetics treated with metformin and diabetics treated with other glucose lowering drugs). CONCLUSION Metformin may lower the risk of AAA repair related mortality and surgical complications among patients with diabetes.
Collapse
Affiliation(s)
- Agnieszka Turowicz
- Dept of Vascular, General and Transplantation Surgery, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland; Dept of Surgery, 4(th) Military Teaching Hospital, Wroclaw, Poland.
| | - Jakub Kobecki
- Dept of Surgery, 4(th) Military Teaching Hospital, Wroclaw, Poland; Division of Oncology and Palliative Care, Department of Clinical Nursing, Faculty of Health Science, Wroclaw Medical University, Wroclaw, Poland
| | | | - Jan Wojciechowski
- Dept of Surgery, 4(th) Military Teaching Hospital, Wroclaw, Poland; Division of Oncology and Palliative Care, Department of Clinical Nursing, Faculty of Health Science, Wroclaw Medical University, Wroclaw, Poland
| | | | - Mariusz Chabowski
- Dept of Surgery, 4(th) Military Teaching Hospital, Wroclaw, Poland; Division of Oncology and Palliative Care, Department of Clinical Nursing, Faculty of Health Science, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
132
|
Golden TN, Simmons RA. Immune dysfunction in developmental programming of type 2 diabetes mellitus. Nat Rev Endocrinol 2021; 17:235-245. [PMID: 33526907 PMCID: PMC7969450 DOI: 10.1038/s41574-020-00464-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2020] [Indexed: 01/30/2023]
Abstract
Intrauterine growth restriction (IUGR) is a common complication of pregnancy and increases the risk of the offspring developing type 2 diabetes mellitus (T2DM) later in life. Alterations in the immune system are implicated in the pathogenesis of IUGR-induced T2DM. The development of the fetal immune system is a delicate balance as it must remain tolerant of maternal antigens whilst also preparing for the post-birth environment. In addition, the fetal immune system is susceptible to an altered intrauterine milieu caused by maternal and placental inflammatory mediators or secondary to nutrient and oxygen deprivation. Pancreatic-resident macrophages populate the pancreas during fetal development, and their phenotype is dynamic through the neonatal period. Furthermore, macrophages in the islets are instrumental in islet development as they influence β-cell proliferation and islet neogenesis. In addition, cytokines, derived from β-cells and macrophages, are important to islet homeostasis in the fetus and adult and, when perturbed, can cause islet dysfunction. Several activated immune pathways have been identified in the islets of people who experienced IUGR, with alternations in the levels of IL-1β and IL-4 as well as changes in TGFβ signalling. Leptin levels are also altered. Immunomodulation has shown therapeutic benefit in T2DM and might be particularly useful in IUGR-induced T2DM.
Collapse
Affiliation(s)
- Thea N Golden
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA.
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA.
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
133
|
Gu Y, Zhang Y, Li M, Huang Z, Jiang J, Chen Y, Chen J, Jia Y, Zhang L, Zhou F. Ferulic Acid Ameliorates Atherosclerotic Injury by Modulating Gut Microbiota and Lipid Metabolism. Front Pharmacol 2021; 12:621339. [PMID: 33841148 PMCID: PMC8026864 DOI: 10.3389/fphar.2021.621339] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a leading cause of death worldwide. Recent studies have emphasized the significance of gut microbiota and lipid metabolism in the development of atherosclerosis. Herein, the effects and molecular mechanisms involving ferulic acid (FA) was examined in atherosclerosis using the ApoE-knockout (ApoE-∕-, c57BL/6 background) mouse model. Eighteen male ApoE-/- mice were fed a high-fat diet (HFD) for 12 weeks and then randomly divided into three groups: the model group, the FA (40 mg/kg/day) group and simvastatin (5 mg/kg/day) group. As results, FA could significantly alleviate atherosclerosis and regulate lipid levels in mice. Liver injury and hepatocyte steatosis induced by HFD were also mitigated by FA. FA improved lipid metabolism involving up-regulation of AMPKα phosphorylation and down-regulation of SREBP1 and ACC1 expression. Furthermore, FA induced marked structural changes in the gut microbiota and fecal metabolites and specifically reduced the relative abundance of Fimicutes, Erysipelotrichaceae and Ileibacterium, which were positively correlated with serum lipid levels in atherosclerosis mice. In conclusion, we demonstrate that FA could significantly ameliorate atherosclerotic injury, which may be partly by modulating gut microbiota and lipid metabolism via the AMPKα/SREBP1/ACC1 pathway.
Collapse
Affiliation(s)
- Yuyan Gu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yaxin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Mei Li
- VIP Healthcare Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiyong Huang
- Department of Otolaryngology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jing Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yihao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Junqi Chen
- Department of Otolaryngology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yuhua Jia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lihua Zhang
- Department of Gynaecology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Fenghua Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
134
|
From Mitochondria to Atherosclerosis: The Inflammation Path. Biomedicines 2021; 9:biomedicines9030258. [PMID: 33807807 PMCID: PMC8000234 DOI: 10.3390/biomedicines9030258] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a key process in metazoan organisms due to its relevance for innate defense against infections and tissue damage. However, inflammation is also implicated in pathological processes such as atherosclerosis. Atherosclerosis is a chronic inflammatory disease of the arterial wall where unstable atherosclerotic plaque rupture causing platelet aggregation and thrombosis may compromise the arterial lumen, leading to acute or chronic ischemic syndromes. In this review, we will focus on the role of mitochondria in atherosclerosis while keeping inflammation as a link. Mitochondria are the main source of cellular energy. Under stress, mitochondria are also capable of controlling inflammation through the production of reactive oxygen species (ROS) and the release of mitochondrial components, such as mitochondrial DNA (mtDNA), into the cytoplasm or into the extracellular matrix, where they act as danger signals when recognized by innate immune receptors. Primary or secondary mitochondrial dysfunctions are associated with the initiation and progression of atherosclerosis by elevating the production of ROS, altering mitochondrial dynamics and energy supply, as well as promoting inflammation. Knowing and understanding the pathways behind mitochondrial-based inflammation in atheroma progression is essential to discovering alternative or complementary treatments.
Collapse
|
135
|
Mbara KC, Mofo Mato PE, Driver C, Nzuza S, Mkhombo NT, Gcwensa SK, Mcobothi EN, Owira PM. Metformin turns 62 in pharmacotherapy: Emergence of non-glycaemic effects and potential novel therapeutic applications. Eur J Pharmacol 2021; 898:173934. [PMID: 33609563 DOI: 10.1016/j.ejphar.2021.173934] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/24/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
Metformin is the most commonly prescribed oral antidiabetic medication. Direct/indirect activation of Adenosine Monophosphate-activated protein kinase (AMPK) and non-AMPK pathways, amongst others, are deemed to explain the molecular mechanisms of action of metformin. Metformin is an established insulin receptor sensitising antihyperglycemic agent, is highly affordable, and has superior safety and efficacy profiles. Emerging experimental and clinical evidence suggests that metformin has pleiotropic non-glycemic effects. Metformin appears to have weight stabilising, renoprotective, neuroprotective, cardio-vascular protective, and antineoplastic effects and mitigates polycystic ovarian syndrome. Anti-inflammatory and antioxidant effects of metformin seem to qualify it as an adjunct therapy in treating infectious diseases such as tuberculosis, viral hepatitis, and the current novel Covid-19 infections. So far, metformin is the only prescription medicine relevant to the emerging field of senotherapeutics. Non-glycemic effects of metformin favourable to its repurposing in therapeutic use are hereby discussed.
Collapse
Affiliation(s)
- Kingsley C Mbara
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Pascale E Mofo Mato
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Christine Driver
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Sanelisiwe Nzuza
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Ntokozo T Mkhombo
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Senamile Kp Gcwensa
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Esethu N Mcobothi
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Peter Mo Owira
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa.
| |
Collapse
|
136
|
Bai B, Chen H. Metformin: A Novel Weapon Against Inflammation. Front Pharmacol 2021; 12:622262. [PMID: 33584319 PMCID: PMC7880161 DOI: 10.3389/fphar.2021.622262] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
It has become widely accepted that inflammation is a driving force behind a variety of chronic diseases, such as cardiovascular disease, diabetes, kidney disease, cancer, neurodegenerative disorders, etc. However, the existing nonsteroidal anti-inflammatory drugs show a limited utility in clinical patients. Therefore, the novel agents with different inflammation-inhibitory mechanisms are worth pursuing. Metformin, a synthetic derivative of guanidine, has a history of more than 50 years of clinical experience in treating patients with type 2 diabetes. Intense research efforts have been dedicated to proving metformin’s inflammation-inhibitory effects in cells, animal models, patient records, and randomized clinical trials. The emerging evidence also indicates its therapeutic potential in clinical domains other than type 2 diabetes. Herein, this article appraises current pre-clinical and clinical findings, emphasizing metformin’s anti-inflammatory properties under individual pathophysiological scenarios. In summary, the anti-inflammatory effects of metformin are evident in pre-clinical models. By comparison, there are still clinical perplexities to be addressed in repurposing metformin to inflammation-driven chronic diseases. Future randomized controlled trials, incorporating better stratification/targeting, would establish metformin’s utility in this clinical setting.
Collapse
Affiliation(s)
- Bo Bai
- Department of Cardiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Haibo Chen
- Department of Cardiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
137
|
Zhu X, Shen W, Liu Z, Sheng S, Xiong W, He R, Zhang X, Ma L, Ju Z. Effect of Metformin on Cardiac Metabolism and Longevity in Aged Female Mice. Front Cell Dev Biol 2021; 8:626011. [PMID: 33585467 PMCID: PMC7877555 DOI: 10.3389/fcell.2020.626011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/09/2020] [Indexed: 12/20/2022] Open
Abstract
The antidiabetic drug metformin exerts pleiotropic effects on multiple organs, including the cardiovascular system. Evidence has shown that metformin improves healthspan and lifespan in male mice, yet its lifespan lengthening effect in females remains elusive. We herein demonstrated that metformin fails to extend the lifespan in female mice. Compared to 2-month-old young controls, 20-month-old female mice showed a spectrum of degenerative cardiac phenotypes alongside significant alterations in the extracellular matrix composition. Despite lowered reactive oxygen species production, long-term metformin treatment did not improve cardiac function in the aged female mice. In contrast, RNA sequencing analyses demonstrated that metformin treatment elevated the extracellular matrix-related gene while lowering oxidative phosphorylation-related gene expression in the heart. In addition, metformin treatment induced metabolic reprogramming that suppressed mitochondrial respiration but activated glycolysis (i.e., Warburg effect) in cultured primary cardiomyocytes and macrophages, thereby sustaining an inflammatory status and lowering ATP production. These findings suggest the unexpected detrimental effects of metformin on the regulation of cardiac homeostasis and longevity in female mice, reinforcing the significance of comprehensive testing prior to the translation of metformin-based novel therapies.
Collapse
Affiliation(s)
- Xudong Zhu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Weiyan Shen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Zhu Liu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Shihao Sheng
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Xiong
- Institute on Aging and Brain Disorders, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ruikun He
- By-Health Co. Ltd., Guangzhou, China
| | | | - Likun Ma
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
138
|
Yang Q, Ma Q, Xu J, Liu Z, Zou J, Shen J, Zhou Y, Da Q, Mao X, Lu S, Fulton DJ, Weintraub NL, Bagi Z, Hong M, Huo Y. Prkaa1 Metabolically Regulates Monocyte/Macrophage Recruitment and Viability in Diet-Induced Murine Metabolic Disorders. Front Cell Dev Biol 2021; 8:611354. [PMID: 33511118 PMCID: PMC7835533 DOI: 10.3389/fcell.2020.611354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/14/2020] [Indexed: 11/13/2022] Open
Abstract
Myeloid cells, including monocytes/macrophages, primarily rely on glucose and lipid metabolism to provide the energy and metabolites needed for their functions and survival. AMP-activated protein kinase (AMPK, its gene is PRKA for human, Prka for rodent) is a key metabolic sensor that regulates many metabolic pathways. We studied recruitment and viability of Prkaa1-deficient myeloid cells in mice and the phenotype of these mice in the context of cardio-metabolic diseases. We found that the deficiency of Prkaa1 in myeloid cells downregulated genes for glucose and lipid metabolism, compromised glucose and lipid metabolism of macrophages, and suppressed their recruitment to adipose, liver and arterial vessel walls. The viability of macrophages in the above tissues/organs was also decreased. These cellular alterations resulted in decreases in body weight, insulin resistance, and lipid accumulation in liver of mice fed with a high fat diet, and reduced the size of atherosclerotic lesions of mice fed with a Western diet. Our results indicate that AMPKα1/PRKAA1-regulated metabolism supports monocyte recruitment and macrophage viability, contributing to the development of diet-induced metabolic disorders including diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Qiuhua Yang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Qian Ma
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jiean Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Zhiping Liu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jianqiu Zou
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jian Shen
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yaqi Zhou
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qingen Da
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaoxiao Mao
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Sarah Lu
- Trinity College of Arts & Sciences, Duke University, Durham, NC, United States
| | - David J. Fulton
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Neal L. Weintraub
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
139
|
Chew GM, Padua AJP, Chow DC, Souza SA, Clements DM, Corley MJ, Pang AP, Alejandria MM, Gerschenson M, Shikuma CM, Ndhlovu LC. Effects of Brief Adjunctive Metformin Therapy in Virologically Suppressed HIV-Infected Adults on Polyfunctional HIV-Specific CD8 T Cell Responses to PD-L1 Blockade. AIDS Res Hum Retroviruses 2021; 37:24-33. [PMID: 33019813 DOI: 10.1089/aid.2020.0172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Targeting inhibitory immune checkpoint receptor pathways has shown remarkable success in improving anticancer T cell responses for the elimination of tumors. Such immunotherapeutic strategies are being pursued for HIV remission. Metformin has shown favorable clinical outcomes in enhancing the efficacy of programmed cell death-1 (PD-1) blockade and restoring antitumor T cell immunity. Furthermore, monocytes are known to be a strong predictor of progression-free survival in response to anti-PD-1 immunotherapy. In a single-arm clinical trial, we evaluated the immunological effects over an 8-week course of metformin therapy in seven euglycemic, virally suppressed HIV-infected participants on combination antiretroviral therapy (cART). We assessed changes in peripheral HIV-Gag-specific T cell responses to immune checkpoint blockade (ICB) with anti-PD-L1 and anti-T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) monoclonal antibodies (mAbs) and changes in CD8 T cell and monocyte subsets using flow cytometry. Study participants were all male, 71% (5/7) Caucasian, with a median age of 61 years, CD4 count of 739 cells/μL, and plasma HIV RNA of <50 copies/mL on stable cART for >1 year. Ex vivo polyfunctional HIV-Gag-specific CD8 T cell responses to anti-PD-L1 mAb significantly improved (p < .05) over the 8-week course of metformin therapy. Moreover, frequencies of both intermediate (CD14+CD16+; r = 0.89, p = .01) and nonclassical (CD14lowCD16+; r = 0.92, p = .01) monocytes at entry were predictive of the magnitude of the anti-HIV CD8 T cell responses to PD-L1 blockade. Collectively, these findings highlight that 8-week course of metformin increases the polyfunctionality of CD8 T cells and that baseline monocyte subset frequencies may be a potential determinant of PD-L1 blockade efficacy. These data provide valuable information for HIV remission trials that utilize ICB strategies to enhance anti-HIV CD8 T cell immunity.
Collapse
Affiliation(s)
- Glen M. Chew
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| | - Ana Joy P. Padua
- College of Medicine, University of the Philippines, Manila, Philippines
| | - Dominic C. Chow
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| | - Scott A. Souza
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| | | | - Michael J. Corley
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| | - Alina P.S. Pang
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| | | | - Mariana Gerschenson
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| | - Cecilia M. Shikuma
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| | - Lishomwa C. Ndhlovu
- John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawaii, USA
| |
Collapse
|
140
|
Feng Y, He M, Ma B, Yang S, Li J, Wen Z, Ouyang H, Zhang W. Therapeutic targets and biological mechanisms of curcumol on atherosclerosis: A study based on network pharmacology approach and biological studies. Pharmacogn Mag 2021. [DOI: 10.4103/pm.pm_336_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
141
|
Cytokine Storm and Failed Resolution in COVID-19: Taking a Cue from Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1352:211-222. [PMID: 35132603 PMCID: PMC9798045 DOI: 10.1007/978-3-030-85109-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Excessive inflammatory responses and failed resolution are major common causes of tissue injury and organ dysfunction in a variety of diseases, including multiple sclerosis (MS), diabetes, and most recently, COVID-19, despite the distinct pathoetiology of the diseases. The promotion of the natural process of inflammatory resolution has been long recognized to improve functional recovery and disease outcomes effectively. To mitigate the excessive inflammation in MS, scientific investigations identified a group of derivatives of omega fatty acids, known as specialized pro-resolving lipid mediators (SPM) that have been significantly effective in treating preclinical disease models of MS. METHODS This chapter is based on our observations from MS. It is being increasingly deliberated that the ongoing COVID-19 infection induces severe cytokine storm that ultimately triggers rampant inflammation. The impact of infection and associated mortality is much higher in patients with co-morbid diseases. Also, reports suggest a better outcome in diabetic patients with reasonable glycemic control, which certainly hints towards a hidden role of anti-hyperglycemic drugs such as metformin in alleviating disease pathology through its anti-inflammatory feature. Notably, SPM and metformin share common therapeutic features in exerting a broad-spectrum anti-inflammatory activity in human patients with a superior safety profile. RESULTS When there is an immediate need to encounter the fast-rampant infection of COVID-19 and control the viral-infection associated morbid inflammatory cytokine storm causing severe organ damage, SPM and metformin should be seriously considered as a potential adjunctive treatment. CONCLUSION Given the fact that current treatment for COVID-19 is only supportive, global research is aimed at developing safe and effective therapeutic options that can result in a better clinical course in patients with comorbid conditions. Accordingly, taking a cue from our experiences in controlling robust inflammatory response in MS and diabetes by simultaneously inhibiting inflammatory process and stimulating its resolution, combinatorial therapy of metformin and SPM in COVID-19 holds significant promise in treating this global health crisis.
Collapse
|
142
|
Yuan Z, Heng Z, Lu Y, Wei J, Cai Z. The Protective Effect of Metformin on Abdominal Aortic Aneurysm: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:721213. [PMID: 34394010 PMCID: PMC8355809 DOI: 10.3389/fendo.2021.721213] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/12/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) patients have a lower risk of abdominal aortic aneurysm (AAA) and its comorbidities, which might be associated with the usage of metformin. The objective of the study was to evaluate the role of metformin in the process of AAA development. METHOD PubMed, Embase and Cochrane Library were searched up to May 15th, 2021. We implemented several methods including the risk of bias graph, GRADE system and funnel plot to assess the quality and possible bias of this study. Subgroup analysis and sensitivity analysis were applied to address quality differences and validate the robustness of the final results. RESULT Ten articles were enrolled after screening 151 articles searched from databases. The pooled results showed that, compared with T2DM patients without metformin, metformin prescription was associated with a slower annual growth rate of the aneurysm (mean difference (MD) -0.67 cm [95% confidence interval (CI) -1.20 ~ -0.15 cm]). Besides, metformin exposure was associated with a lower frequency of AAA events (odds ratio (OR) 0.61 [95% CI 0.41-0.92]). CONCLUSION Metformin alleviated both annual expansion rate and aneurysm rupture frequency in AAA patients with T2DM. SYSTEMATIC REVIEW REGISTRATION PROSPERO, identifier https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=217859 (CRD42020217859).
Collapse
Affiliation(s)
- Zhen Yuan
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijian Heng
- Department of Oncology, Hospital of Chinese Medicine of Changxing County, Huzhou, China
| | - Yi Lu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Wei
- Department of Urology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Zhejun Cai, ; Jia Wei,
| | - Zhejun Cai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Zhejun Cai, ; Jia Wei,
| |
Collapse
|
143
|
Salvatore T, Pafundi PC, Galiero R, Rinaldi L, Caturano A, Vetrano E, Aprea C, Albanese G, Di Martino A, Ricozzi C, Imbriani S, Sasso FC. Can Metformin Exert as an Active Drug on Endothelial Dysfunction in Diabetic Subjects? Biomedicines 2020; 9:biomedicines9010003. [PMID: 33375185 PMCID: PMC7822116 DOI: 10.3390/biomedicines9010003] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular mortality is a major cause of death among in type 2 diabetes (T2DM). Endothelial dysfunction (ED) is a well-known important risk factor for the development of diabetes cardiovascular complications. Therefore, the prevention of diabetic macroangiopathies by preserving endothelial function represents a major therapeutic concern for all National Health Systems. Several complex mechanisms support ED in diabetic patients, frequently cross-talking each other: uncoupling of eNOS with impaired endothelium-dependent vascular response, increased ROS production, mitochondrial dysfunction, activation of polyol pathway, generation of advanced glycation end-products (AGEs), activation of protein kinase C (PKC), endothelial inflammation, endothelial apoptosis and senescence, and dysregulation of microRNAs (miRNAs). Metformin is a milestone in T2DM treatment. To date, according to most recent EASD/ADA guidelines, it still represents the first-choice drug in these patients. Intriguingly, several extraglycemic effects of metformin have been recently observed, among which large preclinical and clinical evidence support metformin’s efficacy against ED in T2DM. Metformin seems effective thanks to its favorable action on all the aforementioned pathophysiological ED mechanisms. AMPK pharmacological activation plays a key role, with metformin inhibiting inflammation and improving ED. Therefore, aim of this review is to assess metformin’s beneficial effects on endothelial dysfunction in T2DM, which could preempt development of atherosclerosis.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio 7, I-80138 Naples, Italy;
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Concetta Aprea
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Carmen Ricozzi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Simona Imbriani
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
- Correspondence: ; Tel.: +39-081-566-5010
| |
Collapse
|
144
|
Hu DJ, Li ZY, Zhu YT, Li CC. Overexpression of long noncoding RNA ANRIL inhibits phenotypic switching of vascular smooth muscle cells to prevent atherosclerotic plaque development in vivo. Aging (Albany NY) 2020; 13:4299-4316. [PMID: 33411680 PMCID: PMC7906209 DOI: 10.18632/aging.202392] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/05/2020] [Indexed: 01/30/2023]
Abstract
Background: Phenotypic switching of vascular smooth muscle cells (VSMCs) plays a key role in atherosclerosis. Long noncoding RNA ANRIL (lncRNA-ANRIL) is critical in vascular homeostasis. Metformin produces multiple beneficial effects in atherosclerosis. However, the underlying mechanisms need to be elucidated. Methods and Results: Metformin increased lncRNA-ANRIL expression and AMPK activity in cultured VSMCs, and inhibited the phenotypic switching of VSMCs to the synthetic phenotype induced by platelet-derived growth factor (PDGF). Overexpression of lncRNA-ANRIL inhibited phenotypic switching and reversed the reduction of AMPK activity in PDGF-treated VSMCs. While, gene knockdown of lncRNA-ANRIL by adenovirus or silence of AMPKγ through siRNA abolished AMPK activation induced by metformin in VSMCs. RNA-immunoprecipitation analysis indicated that the affinity of lncRNA-ANRIL to AMPKγ subunit was increased by metformin. In vivo, administration of metformin increased the levels of lncRNA-ANRIL, suppressed VSMC phenotypic switching, and prevented the development of atherosclerotic plaque in Apoe-/- mice fed with western diet. These protective effects of metformin were abolished by infecting Apoe-/- mice with adenovirus expressing lncRNA-ANRIL shRNA. The levels of AMPK phosphorylation, AMPK activity, and lncRNA-ANRIL expression were decreased in human atherosclerotic lesions. Conclusion: Metformin activates AMPK to suppress the formation of atherosclerotic plaque through upregulation of lncRNA-ANRIL.
Collapse
Affiliation(s)
- Da-Jun Hu
- Department of Cardiology, The First People's Hospital of Chenzhou, Chenzhou 423000, China
| | - Zhen-Yu Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuan-Ting Zhu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chuan-Chang Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
145
|
Sciannimanico S, Grimaldi F, Vescini F, De Pergola G, Iacoviello M, Licchelli B, Guastamacchia E, Giagulli VA, Triggiani V. Metformin: Up to Date. Endocr Metab Immune Disord Drug Targets 2020; 20:172-181. [PMID: 31670618 DOI: 10.2174/1871530319666190507125847] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Metformin is an oral hypoglycemic agent extensively used as first-line therapy for type 2 diabetes. It improves hyperglycemia by suppressing hepatic glucose production and increasing glucose uptake in muscles. Metformin improves insulin sensitivity and shows a beneficial effect on weight control. Besides its metabolic positive effects, Metformin has direct effects on inflammation and can have immunomodulatory and antineoplastic properties. AIM The aim of this narrative review was to summarize the up-to-date evidence from the current literature about the metabolic and non-metabolic effects of Metformin. METHODS We reviewed the current literature dealing with different effects and properties of Metformin and current recommendations about the use of this drug. We identified keywords and MeSH terms in Pubmed and the terms Metformin and type 2 diabetes, type 1 diabetes, pregnancy, heart failure, PCOS, etc, were searched, selecting only significant original articles and review in English, in particular of the last five years. CONCLUSION Even if many new effective hypoglycemic agents have been launched in the market in the last few years, Metformin would always keep a place in the treatment of type 2 diabetes and its comorbidities because of its multiple positive effects and low cost.
Collapse
Affiliation(s)
| | - Franco Grimaldi
- Endocrinology and Metabolism Unit, University Hospital of Udine, Udine, Italy
| | - Fabio Vescini
- Endocrinology and Metabolism Unit, University Hospital of Udine, Udine, Italy
| | - Giovanni De Pergola
- Clinical Nutrition Unit, Medical Oncology, Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Massimo Iacoviello
- University Cardiology Unit, Cardiothoracic Department, Policlinic University Hospital, Bari, Italy
| | - Brunella Licchelli
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Vito A Giagulli
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| |
Collapse
|
146
|
Lima FR, Ferreira LDM, Malta TA, Bonyek-Silva I, Santos RL, Tavares NM, de Carvalho Filho EM, Arruda S. Metformin promotes susceptibility to experimental Leishmania braziliensis infection. Mem Inst Oswaldo Cruz 2020; 115:e200272. [PMID: 33206822 PMCID: PMC7668079 DOI: 10.1590/0074-02760200272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/03/2020] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Metformin (MET) is a hypoglycemic drug used for the treatment of diabetes, despite interference in host immunity against microorganisms. Cutaneous infection caused by pathogens such as Leishmania braziliensis (Lb), the agent responsible for cutaneous leishmaniasis (CL) in Brazil, represents an interesting model in which to evaluate the effects associated with MET. OBJECTIVE To evaluate the modulatory effect of MET in Lb infection. MATERIAL AND METHODS Experimental study of Lb infection and MET treatment in BALB/c mice and Raw 264.7 macrophages. FINDINGS MET treatment interfered with lesion kinetics, increased parasite load and reduced macrophage proliferation. Low concentrations of MET in Lb culture allow for the maintenance of stationary parasite growth phase. Lb-infected cells treated with MET exhibited increased parasite load. While both MET and Lb infection alone promoted the production of intracellular reactive oxygen species (ROS), reduced levels of ROS were seen in MET-treated Lb-infected macrophages. MAIN CONCLUSION Experimental treatment with MET interfered with the kinetics of cutaneous ulceration, increased Lb parasite load, altered ROS production and modulated cellular proliferation. Our experimental results indicate that MET interfere with the evolution of CL.
Collapse
Affiliation(s)
- Filipe Rocha Lima
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, BA, Brasil
- Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Lais de Melo Ferreira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, BA, Brasil
- Universidade Estadual da Bahia, Departamento de Ciências da Vida, Salvador, BA, Brasil
| | - Tainá Alves Malta
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, BA, Brasil
- Universidade Estadual da Bahia, Departamento de Ciências da Vida, Salvador, BA, Brasil
| | - Icaro Bonyek-Silva
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório de Interação Parasito-Hospedeiro e Epidemiologia, Salvador, BA, Brasil
- Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Reinan Lima Santos
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório de Interação Parasito-Hospedeiro e Epidemiologia, Salvador, BA, Brasil
- Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Natália Machado Tavares
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório de Interação Parasito-Hospedeiro e Epidemiologia, Salvador, BA, Brasil
| | - Edgar Marcelino de Carvalho Filho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório de Pesquisa Clínica, Salvador, BA, Brasil
- Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Sérgio Arruda
- Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, BA, Brasil
- Universidade Estadual da Bahia, Departamento de Ciências da Vida, Salvador, BA, Brasil
| |
Collapse
|
147
|
Robb JL, Morrissey NA, Weightman Potter PG, Smithers HE, Beall C, Ellacott KLJ. Immunometabolic Changes in Glia - A Potential Role in the Pathophysiology of Obesity and Diabetes. Neuroscience 2020; 447:167-181. [PMID: 31765625 PMCID: PMC7567742 DOI: 10.1016/j.neuroscience.2019.10.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
Chronic low-grade inflammation is a feature of the pathophysiology of obesity and diabetes in the CNS as well as peripheral tissues. Glial cells are critical mediators of the response to inflammation in the brain. Key features of glia include their metabolic flexibility, sensitivity to changes in the CNS microenvironment, and ability to rapidly adapt their function accordingly. They are specialised cells which cooperate to promote and preserve neuronal health, playing important roles in regulating the activity of neuronal networks across the brain during different life stages. Increasing evidence points to a role of glia, most notably astrocytes and microglia, in the systemic regulation of energy and glucose homeostasis in the course of normal physiological control and during disease. Inflammation is an energetically expensive process that requires adaptive changes in cellular metabolism and, in turn, metabolic intermediates can also have immunomodulatory actions. Such "immunometabolic" changes in peripheral immune cells have been implicated in contributing to disease pathology in obesity and diabetes. This review will discuss the evidence for a role of immunometabolic changes in glial cells in the systemic regulation of energy and glucose homeostasis, and how this changes in the context of obesity and diabetes.
Collapse
Affiliation(s)
- Josephine L Robb
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Nicole A Morrissey
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Paul G Weightman Potter
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Hannah E Smithers
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
148
|
Metformin and Systemic Metabolism. Trends Pharmacol Sci 2020; 41:868-881. [PMID: 32994049 DOI: 10.1016/j.tips.2020.09.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/14/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022]
Abstract
Metformin can improve patients' hyperglycemia through significant suppression of hepatic glucose production. However, up to 300 times higher concentrations of metformin accumulate in the intestine than in the circulation, where it alters nutrient metabolism in intestinal epithelial cells and microbiome, leading to increased lactate production. Hepatocytes use lactate to make glucose at the cost of energy expenditure, creating a futile intestine-liver cycle. Furthermore, metformin reduces blood lipopolysaccharides and its initiated low-grade inflammation and increased oxidative phosphorylation in liver and adipose tissues. These metformin effects result in the improvement of insulin sensitivity and glucose utilization in extrahepatic tissues. In this review, I discuss the current understanding of the impact of metformin on systemic metabolism and its molecular mechanisms of action in various tissues.
Collapse
|
149
|
Canagliflozin alleviates LPS-induced acute lung injury by modulating alveolar macrophage polarization. Int Immunopharmacol 2020; 88:106969. [PMID: 33182027 DOI: 10.1016/j.intimp.2020.106969] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/31/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Canagliflozin (CANA), a sodium-glucose cotransporter 2 inhibitor, is a novel therapeutic agent that exhibits multiple actions in type 2 diabetes. CANA can regulate intracellular glucose metabolism and exert anti-inflammatory effects in immune cells. Alveolar macrophage polarization balance is often associated with lower inflammation in acute lung injury (ALI). However, little is known about the anti-inflammatory effect of CANA on ALI. METHODS This study aimed to determine the effect of CANA on ALI as well as its potential ability to modulate alveolar macrophage polarization in ALI mouse models and bone marrow-derived macrophages (BMDMs). RESULTS The histopathological changes indicated that CANA alleviated lung injury in lipopolysaccharide-induced ALI mice models and exerted anti-inflammatory effects in the presence of lower levels of tumor necrosis factor-ɑ, interleukin-6, and interleukin-1β in bronchoalveolar lavage fluid (BALF) and serum. Moreover, flow cytometry analysis of mouse BALF cells and BMDMs demonstrated that CANA can modulate and reconstitute M1 and M2 macrophage balance, inhibiting macrophages with the M1 phenotype while promoting macrophages to shift to the M2 phenotype. Immunohistochemistry and reverse transcription polymerase chain reaction were also performed. CONCLUSIONS These findings indicate that CANA alleviates lung injury and exerts anti-inflammatory effects by modulating alveolar macrophage polarization balance, suggesting that CANA might act as a novel anti-inflammatory drug for treating ALI.
Collapse
|
150
|
Kong G, Zhou L, Serger E, Palmisano I, De Virgiliis F, Hutson TH, Mclachlan E, Freiwald A, La Montanara P, Shkura K, Puttagunta R, Di Giovanni S. AMPK controls the axonal regenerative ability of dorsal root ganglia sensory neurons after spinal cord injury. Nat Metab 2020; 2:918-933. [PMID: 32778834 DOI: 10.1038/s42255-020-0252-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/26/2020] [Indexed: 12/25/2022]
Abstract
Regeneration after injury occurs in axons that lie in the peripheral nervous system but fails in the central nervous system, thereby limiting functional recovery. Differences in axonal signalling in response to injury that might underpin this differential regenerative ability are poorly characterized. Combining axoplasmic proteomics from peripheral sciatic or central projecting dorsal root ganglion (DRG) axons with cell body RNA-seq, we uncover injury-dependent signalling pathways that are uniquely represented in peripheral versus central projecting sciatic DRG axons. We identify AMPK as a crucial regulator of axonal regenerative signalling that is specifically downregulated in injured peripheral, but not central, axons. We find that AMPK in DRG interacts with the 26S proteasome and its CaMKIIα-dependent regulatory subunit PSMC5 to promote AMPKα proteasomal degradation following sciatic axotomy. Conditional deletion of AMPKα1 promotes multiple regenerative signalling pathways after central axonal injury and stimulates robust axonal growth across the spinal cord injury site, suggesting inhibition of AMPK as a therapeutic strategy to enhance regeneration following spinal cord injury.
Collapse
Affiliation(s)
- Guiping Kong
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Graduate School for Cellular and Molecular Neuroscience, University of Tübingen, Tübingen, Germany
| | - Luming Zhou
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Graduate School for Cellular and Molecular Neuroscience, University of Tübingen, Tübingen, Germany
| | - Elisabeth Serger
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Ilaria Palmisano
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Francesco De Virgiliis
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Thomas H Hutson
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Eilidh Mclachlan
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Anja Freiwald
- Proteomics Core Facility, Institute of Molecular Biology, Mainz, Germany
| | - Paolo La Montanara
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Kirill Shkura
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Radhika Puttagunta
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- University of Heidelberg, Heidelberg, Germany
| | - Simone Di Giovanni
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK.
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| |
Collapse
|