51
|
Suzuki T, Yamashita S, Hattori K, Matsuda N, Hattori Y. Impact of a long-term high-glucose environment on pro-inflammatory responses in macrophages stimulated with lipopolysaccharide. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:2129-2139. [PMID: 34402957 DOI: 10.1007/s00210-021-02137-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 11/27/2022]
Abstract
Cumulative evidence has established that macrophages orchestrate inflammatory responses that crucially contribute to the pathogenesis of insulin-resistant obesity and type 2 diabetes. In the present study, we examined the impact of hyperglycemia on macrophage pro-inflammatory responses under an inflammatory stimulus. To conduct this study, RAW264.7 macrophages were cultured under normal- (5.5 mM) or high-glucose (22 or 40 mM) conditions for 7 days and stimulated with lipopolysaccharide (LPS). Long-term exposure to high glucose significantly enhanced the increase in the production of pro-inflammatory cytokines, including tumor necrosis-α, interleukin (IL)-1β, and IL-6, when macrophages were stimulated with LPS. The LPS-induced increases in inducible nitric oxide (NO) synthase (iNOS) expression and NO production were also significantly enhanced by long-term exposure of macrophages to high glucose. Treatment with N-acetyl-L-cysteine, a widely used thiol-containing antioxidant, blunted the enhancement of the LPS-induced upregulation of pro-inflammatory cytokine production, iNOS expression, and NO production in macrophages. When intracellular reactive oxygen species (ROS) were visualized using the fluorescence dye 5-(and-6)-chloromethyl-2',7'-dichlorofluorescein diacetate, acetyl ester, a significant increase in ROS generation was found after stimulation of macrophages with LPS, and this increased ROS generation was exacerbated under long-term high-glucose conditions. LPS-induced translocation of phosphorylated nuclear factor-κB (NF-κB), a transcription factor regulating many pro-inflammatory genes, into the nucleus was promoted under long-term high-glucose conditions. Altogether, the present results indicate that a long-term high-glucose environment can enhance activation of NF-κB in LPS-stimulated macrophages possibly due to excessive ROS production, thereby leading to increased macrophage pro-inflammatory responses.
Collapse
Affiliation(s)
- Tokiko Suzuki
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
- J-Pharma Co., Ltd., Yokohama, 230-0046, Japan
| | - Shigeyuki Yamashita
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
- Department of Thoracic and Cardiovascular Surgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Kohshi Hattori
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, 113-8655, Japan
| | - Naoyuki Matsuda
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yuichi Hattori
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Tobetsu, 061-0293, Japan.
| |
Collapse
|
52
|
Wang S, Dai Y. Roles of AMPK and Its Downstream Signals in Pain Regulation. Life (Basel) 2021; 11:life11080836. [PMID: 34440581 PMCID: PMC8401922 DOI: 10.3390/life11080836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/06/2021] [Accepted: 08/12/2021] [Indexed: 12/20/2022] Open
Abstract
Pain is an unpleasant sensory and emotional state that decreases quality of life. A metabolic sensor, adenosine monophosphate-activated protein kinase (AMPK), which is ubiquitously expressed in mammalian cells, has recently attracted interest as a new target of pain research. Abnormal AMPK expression and function in the peripheral and central nervous systems are associated with various types of pain. AMPK and its downstream kinases participate in the regulation of neuron excitability, neuroinflammation and axonal and myelin regeneration. Numerous AMPK activators have reduced pain behavior in animal models. The current understanding of pain has been deepened by AMPK research, but certain issues, such as the interactions of AMPK at each step of pain regulation, await further investigation. This review examines the roles of AMPK and its downstream kinases in neurons and non-neuronal cells, as well as their contribution to pain regulation.
Collapse
Affiliation(s)
- Shenglan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe 650-8530, Japan
- Correspondence: (S.W.); (Y.D.); Tel.: +86-10-53912197 (S.W.); +81-78-304-3147 (Y.D.)
| | - Yi Dai
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe 650-8530, Japan
- Traditional Medicine Research Center, Chinese Medicine Confucius Institute, Hyogo College of Medicine, Kobe 663-8501, Japan
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya 663-8501, Japan
- Correspondence: (S.W.); (Y.D.); Tel.: +86-10-53912197 (S.W.); +81-78-304-3147 (Y.D.)
| |
Collapse
|
53
|
Hemed-Shaked M, Cowman MK, Kim JR, Huang X, Chau E, Ovadia H, Amar KO, Eshkar-Sebban L, Melamed M, Lev LB, Kedar E, Armengol J, Alemany J, Beyth S, Okon E, Kanduc D, Elgavish S, Wallach-Dayan SB, Cohen SJ, Naor D. MTADV 5-MER peptide suppresses chronic inflammations as well as autoimmune pathologies and unveils a new potential target-Serum Amyloid A. J Autoimmun 2021; 124:102713. [PMID: 34390919 DOI: 10.1016/j.jaut.2021.102713] [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: 06/09/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/17/2022]
Abstract
Despite the existence of potent anti-inflammatory biological drugs e.g., anti-TNF and anti IL-6 receptor antibodies, for treating chronic inflammatory and autoimmune diseases, these are costly and not specific. Cheaper oral available drugs remain an unmet need. Expression of the acute phase protein Serum Amyloid A (SAA) is dependent on release of pro-inflammatory cytokines IL-1, IL-6 and TNF-α during inflammation. Conversely, SAA induces pro-inflammatory cytokine secretion, including Th17, leading to a pathogenic vicious cycle and chronic inflammation. 5- MER peptide (5-MP) MTADV (methionine-threonine-alanine-aspartic acid-valine), also called Amilo-5MER, was originally derived from a sequence of a pro-inflammatory CD44 variant isolated from synovial fluid of a Rheumatoid Arthritis (RA) patient. This human peptide displays an efficient anti-inflammatory effects to ameliorate pathology and clinical symptoms in mouse models of RA, Inflammatory Bowel Disease (IBD) and Multiple Sclerosis (MS). Bioinformatics and qRT-PCR revealed that 5-MP, administrated to encephalomyelytic mice, up-regulates genes contributing to chronic inflammation resistance. Mass spectrometry of proteins that were pulled down from an RA synovial cell extract with biotinylated 5-MP, showed that it binds SAA. 5-MP disrupted SAA assembly, which is correlated with its pro-inflammatory activity. The peptide MTADV (but not scrambled TMVAD) significantly inhibited the release of pro-inflammatory cytokines IL-6 and IL-1β from SAA-activated human fibroblasts, THP-1 monocytes and peripheral blood mononuclear cells. 5-MP suppresses the pro-inflammatory IL-6 release from SAA-activated cells, but not from non-activated cells. 5-MP could not display therapeutic activity in rats, which are SAA deficient, but does inhibit inflammations in animal models of IBD and MS, both are SAA-dependent, as shown by others in SAA knockout mice. In conclusion, 5-MP suppresses chronic inflammation in animal models of RA, IBD and MS, which are SAA-dependent, but not in animal models, which are SAA-independent.
Collapse
Affiliation(s)
- Maayan Hemed-Shaked
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Mary K Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, USA
| | - Jin Ryoun Kim
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Xiayun Huang
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Edward Chau
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Haim Ovadia
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Keren-Or Amar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Lora Eshkar-Sebban
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Michal Melamed
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Libat Bar Lev
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Eli Kedar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | - Shaul Beyth
- Orthopedic Surgery Department, Hadassah University Hospital, Jerusalem, Israel
| | - Eli Okon
- Department of Pathology, Hadassah University Hospital, Jerusalem, Israel
| | - Darja Kanduc
- Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70126, Italy
| | - Sharona Elgavish
- Bioinformatics Unit of the Hebrew University of Jerusalem and Hadassah Medical Center, Israel
| | - Shulamit B Wallach-Dayan
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Israel
| | - Shmuel Jaffe Cohen
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - David Naor
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| |
Collapse
|
54
|
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
|
55
|
Liu Z, Liao W, Zhang Z, Sun R, Luo Y, Chen Q, Li X, Lu R, Ying Y. Metformin Affects Gut Microbiota Composition and Diversity Associated with Amelioration of Dextran Sulfate Sodium-Induced Colitis in Mice. Front Pharmacol 2021; 12:640347. [PMID: 34122067 PMCID: PMC8191634 DOI: 10.3389/fphar.2021.640347] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/10/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Inflammatory bowel disease (IBD) is an increasingly common and globally emergent immune-mediated disorder. The etiology of IBD is complex, involving multiple factors such as immune dysregulation, environmental factors, genetic mutations, and microbiota dysbiosis, exacerbated by a lack of effective clinical therapies. Recently, studies hypothesized that dysbiosis of intestinal flora might participate in the onset of IBD. Metformin is widely used to treat type 2 diabetes and has shown beneficial effects in mouse models of IBD, although its underlying mechanisms remain poorly understood. Accumulating studies found that metformin shows beneficial effects for diabetes by affecting microbiota composition. This study explores possible regulatory effects of metformin on intestinal microecology during treatment for IBD. Methods: Inflammation was induced using 3% Dextran Sulfate Sodium (DSS) solution to generate mice models of IBD. Metformin treatments were assayed by measuring body weights and colon lengths of mice and H&E staining to observe histological effects on colon tissue structures. Changes in bacterial community composition and diversity-related to IBD and metformin treatment were assessed by high-throughput metagenomic sequencing analysis. Results: Metformin administration significantly ameliorated body weight loss, inhibited colon shrinking, and contributed to preserving the integrity of colon histological structures. The gut microbiota profiles revealed that the biodiversity of intestinal flora lost during inflammation was restored under metformin treatment. Metformin administration was also associated with decreased pathogenic Escherichia shigella and increased abundance of Lactobacillus and Akkermansia. Conclusion: Metformin appears to induce anti-inflammatory effects, thus ameliorating colitis symptoms, concurrent with enrichment for beneficial taxa and restored microbial diversity, suggesting a viable strategy against IBD.
Collapse
Affiliation(s)
- Zhiyi Liu
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Queen Mary School, Nanchang University, Nanchang, China
| | - Wangdi Liao
- Departments of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zihan Zhang
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Queen Mary School, Nanchang University, Nanchang, China
| | - Ruipu Sun
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Queen Mary School, Nanchang University, Nanchang, China
| | - Yunfei Luo
- Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Qiongfeng Chen
- Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Xin Li
- Departments of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ruiling Lu
- Departments of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Ying
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang, China.,The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
56
|
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
|
57
|
Nozu T, Miyagishi S, Ishioh M, Takakusaki K, Okumura T. Phlorizin attenuates visceral hypersensitivity and colonic hyperpermeability in a rat model of irritable bowel syndrome. Biomed Pharmacother 2021; 139:111649. [PMID: 33957565 DOI: 10.1016/j.biopha.2021.111649] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
Visceral hypersensitivity and impaired gut barrier are crucial contributors to the pathophysiology of irritable bowel syndrome (IBS), and those are mediated via corticotropin-releasing factor (CRF)-Toll like receptor 4-pro-inflammatory cytokine signaling. Phlorizin is an inhibitor of sodium-linked glucose transporters (SGLTs), and known to have anti-cytokine properties. Thus, we hypothesized that phlorizin may improve these gastrointestinal changes in IBS, and tested this hypothesis in rat IBS models, i.e., lipopolysaccharide (LPS) or CRF-induced visceral hypersensitivity and colonic hyperpermeability. The visceral pain threshold in response to colonic balloon distention was estimated by abdominal muscle contractions by electromyogram, and colonic permeability was measured by quantifying the absorbed Evans blue in colonic tissue. Subcutaneous (s.c.) injection of phlorizin inhibited visceral hypersensitivity and colonic hyperpermeability induced by LPS in a dose-dependent manner. Phlorizin also blocked CRF-induced these gastrointestinal changes. Phlorizin is known to inhibit both SGLT1 and SGLT2, but intragastric administration of phlorizin may only inhibit SGLT1 because gut mainly expresses SGLT1. We found that intragastric phlorizin did not display any effects, but ipragliflozin, an orally active and selective SGLT2 inhibitor improved the gastrointestinal changes in the LPS model. Compound C, an adenosine monophosphate-activated protein kinase (AMPK) inhibitor, NG-nitro-L-arginine methyl ester, a nitric oxide (NO) synthesis inhibitor and naloxone, an opioid receptor antagonist reversed the effects of phlorizin. In conclusions, phlorizin improved visceral hypersensitivity and colonic hyperpermeability in IBS models. These effects may result from inhibition of SGLT2, and were mediated via AMPK, NO and opioid pathways. Phlorizin may be effective for the treatment of IBS.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Center for Medical Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Saori Miyagishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Masatomo Ishioh
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Kaoru Takakusaki
- Division of Neuroscience, Department of Physiology, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Toshikatsu Okumura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| |
Collapse
|
58
|
Davanzo GG, Castro G, Moraes-Vieira PMM. Immunometabolic regulation of adipose tissue resident immune cells. Curr Opin Pharmacol 2021; 58:44-51. [PMID: 33878567 DOI: 10.1016/j.coph.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/19/2022]
Abstract
Adipose tissue (AT) performs immunoregulatory functions beyond fat storage. In addition to adipocytes, AT has a diverse spectrum of resident and infiltrating immune cells in health and disease. Immune cells contribute to the homeostatic function of AT by adapting their metabolism in accordance with the microenvironment. However, how the metabolic reprogramming of immune cells affects their inflammatory profile and the subsequent implication for adipocyte function is not completely elucidated. Here, we discuss the available data on metabolic regulatory processes implicated in the control of adipose tissue-resident immune cells and their crosstalk with adipocytes.
Collapse
Affiliation(s)
- Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil
| | - Gisele Castro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Pedro Manoel M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil; Obesity and Comorbidities Research Center (OCRC), University of Campinas, SP, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, SP, Brazil.
| |
Collapse
|
59
|
The AMPK modulator metformin as adjunct to methotrexate in patients with rheumatoid arthritis: A proof-of-concept, randomized, double-blind, placebo-controlled trial. Int Immunopharmacol 2021; 95:107575. [PMID: 33773207 DOI: 10.1016/j.intimp.2021.107575] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/22/2021] [Accepted: 03/07/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Metformin (MET) may exert anti-rheumatic effects and reduce cartilage degradation through its immunomodulatory and anti-inflammatory actions. METHODS This was a double-blind placebo-controlled study, 120 adult patients with active rheumatoid arthritis (RA) were randomized to receive MET (1000 mg) or placebo daily with methotrexate (MTX, 7.5 mg/week) for 12 weeks. American College of Rheumatology (ACR)20, ACR50, and ACR70 response rates, Disease Activity Score in 28 joints (DAS-28), and drug safety were the efficacy endpoints. Serum levels of TNF-α, IL-1β, IL-6, IL-10, IL-17A, NF-κB, TGG-β1, MDA together with gene expression of AMPK and IGF-IR were assessed before and after the therapy. RESULTS A total of 80.8% of the patients in the MET group, compared with 54.7% in placebo group, met the criteria of ACR20 response after 12 weeks (P = 0.001). Statistically significant enhancements in the DAS28-3 (CRP) were observed after 4 and 8 weeks for the MET group compared with placebo and were sustained after 12 weeks. MET group showed statistically significant increase in percentage of patients achieving DAS remission after 12 weeks (P = 0.015). Significant improvements in ACR50, ACR70, Health Assessment Questionnaire Disability Index (HAQ-DI), and DAS28-3 (CRP) were also reported. MET was well-tolerated, and no serious adverse effects were reported in both groups. Furthermore, the MET group was superior in improving the measured parameters compared to the placebo. CONCLUSIONS MET improved the anti-rheumatic effect of MTX; suggesting it to be a beneficial adjuvant in patients with RA. Trial registration ID: NCT04068246.
Collapse
|
60
|
Leng W, Jiang J, Chen B, Wu Q. Metformin and Malignant Tumors: Not Over the Hill. Diabetes Metab Syndr Obes 2021; 14:3673-3689. [PMID: 34429626 PMCID: PMC8380287 DOI: 10.2147/dmso.s326378] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022] Open
Abstract
Malignant tumors are a major cause of death, and their incidence is increasing worldwide. Although the survival rate for some cancers has improved, treatments for other malignant tumors are limited, and their mortality rate continues to increase. People with type 2 diabetes have a higher risk of malignant tumors and a higher mortality rate than those without diabetes. Metformin is a commonly used hypoglycemic drug. In recent years, a growing number of studies have indicated that metformin has antitumor effects and increases the sensitivity of malignant tumors to chemotherapy. However, the effect of metformin on different tumors is currently controversial, and the mechanism of metformin's antitumor action is not fully understood. Insights into the effect of metformin on malignant tumors and the possible mechanism may contribute to the development of antitumor drugs.
Collapse
Affiliation(s)
- Weiling Leng
- Endocrinology Department, The First Affiliated Hospital of the Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
| | - Juan Jiang
- Endocrinology and Nephrology Department, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, People’s Republic of China
| | - Bing Chen
- Endocrinology Department, The First Affiliated Hospital of the Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China
- Bing Chen Endocrinology Department, The First Affiliated Hospital of the Third Military Medical University (Army Medical University), Chongqing, People’s Republic of China Email
| | - Qinan Wu
- Endocrinology Department, Dazu Hospital of Chongqing Medical University, The People’s Hospital of Dazu, Chongqing, People’s Republic of China
- Correspondence: Qinan Wu Endocrinology Department, Dazu Hospital of Chongqing Medical University, The People’s Hospital of Dazu, Chongqing, People’s Republic of China Email
| |
Collapse
|
61
|
Didari T, Hassani S, Baeeri M, Navaei-Nigjeh M, Rahimifard M, Haghi-Aminjan H, Gholami M, Nejad SM, Hassan FI, Mojtahedzadeh M, Abdollahi M. Short-term Effects of Metformin on Cardiac and Peripheral Blood Cells Following Cecal Ligation and Puncture-induced Sepsis. Drug Res (Stuttg) 2020; 71:257-264. [PMID: 33348389 DOI: 10.1055/a-1322-7478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIM OF THE STUDY Sepsis has well-documented inflammatory effects on cardiovascular and blood cells. This study is designed to investigate potential anti-inflammatory effects of metformin on cardiac and blood cells 12 and 24 h following cecal ligation and puncture (CLP)-induced sepsis. METHODS For the purpose of this study, 36 male Wistar rats were divided into six groups: two groups underwent CLP, two groups underwent CLP and received metformin, and two groups only received sham operations. 12 h later, 18 rats (half of rats in each of the three aforementioned groups) were sacrificed and cardiac and blood cells were harvested. Subsequently, 12 h later, the rest of the rats were euthanatized. In all harvested blood and cardiac cells, oxidative stress indicators, antioxidant properties, count of blood cells, neutrophil infiltration, percentage of weight loss and pathological assessment were conducted. RESULTS In our experiment, metformin elevated antioxidant levels, improved function of blood cells and percentage of weight loss. Moreover, in the groups which received metformin, oxidative stress and neutrophil infiltration markers were decreased significantly. Moreover, pathological investigations of cardiac cell injury were reduced in the metformin group. CONCLUSIONS Our findings suggest that in CLP induced sepsis model, metformin can improve the function of blood and cardiac cells through alleviating inflammation, improvement of anti-inflammation properties, and enhancement of blood profile, and all these effects are more pronounced after 24 h in comparison with 12 h after induction of sepsis.
Collapse
Affiliation(s)
- Tina Didari
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Hassani
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Navaei-Nigjeh
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahban Rahimifard
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mahdi Gholami
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Mohammadi Nejad
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Fatima Ismail Hassan
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Mojtahedzadeh
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Clinical Pharmacy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
62
|
Zhang Y, Liu X, Yang L, Zou L. Current Researches, Rationale, Plausibility, and Evidence Gaps on Metformin for the Management of Hypertensive Disorders of Pregnancy. Front Pharmacol 2020; 11:596145. [PMID: 33381040 PMCID: PMC7768035 DOI: 10.3389/fphar.2020.596145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are a group of morbid pregnancy complications, with preeclampsia (PE) being the most common subclassification among them. PE affects 2%–8% of pregnancies globally and threatens maternal and fetal health seriously. However, the only effective treatment of PE to date is the timely termination of pregnancy, albeit with increased perinatal risks. Hence, more emerging therapies for PE management are in urgent need. Originally introduced as the first-line therapy for type 2 diabetes mellitus, metformin (MET) has now been found in clinical trials to significantly reduce the incidence of gestational hypertension and PE in pregnant women with PE-related risks, including but not limited to pregestational diabetes mellitus, gestational diabetes mellitus, polycystic ovary syndrome, or obesity. Additionally, existing clinical data have preliminarily ensured the safety of taking MET during human pregnancies. Relevant lab studies have indicated that the underlying mechanism includes angiogenesis promotion, endothelial protection, anti-inflammatory effects, and particularly protective effects on trophoblast cells against the risk factors, which are beneficial to placental development. Together with its global availability, easy administration, and low cost, MET is expected to be a promising option for the prevention and treatment of PE. Nevertheless, there are still some limitations in current studies, and the design of the relevant research scheme is supposed to be further improved in the future. Herein, we summarize the relevant clinical and experimental researches to discuss the rationale, safety, and feasibility of MET for the management of HDP. At the end of the article, gaps in current researches are proposed. Concretely, experimental MET concentration and PE models should be chosen cautiously. Besides, the clinical trial protocol should be further optimized to evaluate the reduction in the prevalence of PE as a primary endpoint. All of those evidence gaps may be of guiding significance to improve the design of relevant experiments and clinical trials in the future.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
63
|
Effects of inflammatory and anti-inflammatory environments on the macrophage mitochondrial function. Sci Rep 2020; 10:20324. [PMID: 33230189 PMCID: PMC7684315 DOI: 10.1038/s41598-020-77370-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022] Open
Abstract
Mitochondrial response to inflammation is crucial in the metabolic adaptation to infection. This study aimed to explore the mitochondrial response under inflammatory and anti-inflammatory environments, with a focus on the tricarboxylic acid (TCA) cycle. Expression levels of key TCA cycle enzymes and the autophagy-related protein light chain 3b (LC3b) were determined in raw 264.7 cells treated with lipopolysaccharide (LPS) and metformin (Met). Additionally, reactive oxygen species (ROS) levels and mitochondrial membrane potential were assessed using flow cytometry. Moreover, 8-week-old C57BL/6J mice were intraperitoneally injected with LPS and Met to assess the mitochondrial response in vivo. Upon LPS stimulation, the expression of key TCA enzymes, including citrate synthase, α-ketoglutarate dehydrogenase, and isocitrate dehydrogenase 2, and the mitochondrial membrane potential decreased, whereas the levels of LC3b and ROS increased. However, treatment with Met inhibited the reduction of LPS-induced enzyme levels as well as the elevation of LC3b and ROS levels. In conclusion, the mitochondrial TCA cycle is affected by the inflammatory environment, and the LPS-induced effects can be reversed by Met treatment.
Collapse
|
64
|
Al-Disi D, Ansari MGA, Sabico S, Wani K, Hussain SD, Elshafie MM, McTernan P, Al-Daghri NM. High glucose load and endotoxemia among overweight and obese Arab women with and without diabetes: An observational study. Medicine (Baltimore) 2020; 99:e23211. [PMID: 33181703 PMCID: PMC7668447 DOI: 10.1097/md.0000000000023211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dietary intake influences gut microbiota activity. Nevertheless, there is a lack of evidence available that illustrates the acute effects of high glucose meal on metabolic endotoxemia. The present study assessed the acute impact of high glucose meal on endotoxemia and other clinical parameters in Saudi females with varying degrees of glycemia.The subjects were 64 consenting pre-menopausal women, grouped into 3: control [n = 14 lean, non-T2DM, BMI = 22.2 ± 2.2 kg/m]; overweight [n = 16, non-T2DM, BMI = 28.5 ± 1.5 kg/m] and T2DM [n = 34, BMI = 35.2 ± 7.7 kg/m]. After an overnight fast, all subjects were given a standardized high-glucose (75 g) meal. Anthropometrics were taken and blood samples were withdrawn at baseline and postprandial (0, 2 and 4-hours), serum glucose, endotoxin and lipid profile were quantified.At baseline, total cholesterol, LDL-cholesterol, triglycerides and serum glucose levels were significantly higher (P values <.01) whereas significantly lower HDL-cholesterol levels (P < .01) were observed in T2DM subjects compared to other groups. Baseline endotoxin levels were highest in the overweight group (3.2 ± 1.1 mmol/L) as compared to control (2.0 ± 0.5 mmol/L) and T2DM (2.7 ± 1.2 mmol/L) (P = .046). HDL-cholesterol, LDL-cholesterol and triglycerides, significantly decreased in the T2DM group after 2 hours (P values <.05), whereas unremarkable changes observed in other groups. Lastly, endotoxin levels significantly increased only in the overweight group (3.2 ± 1.1 vs 4.2 ± 1.4 mmol/L; P < .05), 4 hours postprandial.High glucose meal elevates endotoxemia only among overweight subjects and impairs dysbiosis.
Collapse
Affiliation(s)
- Dara Al-Disi
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University
| | | | - Shaun Sabico
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Kaiser Wani
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Syed Danish Hussain
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mona M. Elshafie
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University
| | - Philip McTernan
- School of Science and Technology, Department of Biosciences, Nottingham Trent University, Nottingham, NG1 8NS, UK
| | - Nasser M. Al-Daghri
- Riyadh Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
65
|
Wang M, Qu S, Ma J, Wang X, Yang Y. Metformin Suppresses LPS-Induced Inflammatory Responses in Macrophage and Ameliorates Allergic Contact Dermatitis in Mice via Autophagy. Biol Pharm Bull 2020; 43:129-137. [PMID: 31902918 DOI: 10.1248/bpb.b19-00689] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Allergic contact dermatitis (ACD) is one of the most common skin diseases caused by hapten-modified proteins. Metformin, a drug commonly prescribed for type II diabetes, has been demonstrated to have various biological functions beyond its antidiabetic effects. However, its role in ACD remains unknown. In the present study, we found that metformin reduced the production of nitric oxide (NO) and the level of proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. These anti-inflammatory effects were also demonstrated on bone marrow-derived macrophages (BMDMs). Furthermore, metformin also enhanced autophagic flux, inhibited the phosphorylation of the serine/threonine protein kinase (AKT)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinases (MAPKs) related protein levels and the level of miR-221 in LPS-stimulated RAW264.7 cells. Besides, metformin attenuated 2,4-dinitrofluorobenzene (DNFB)-induced ACD and inhibited proinflammatory cytokines in the ear. In addition, metformin ameliorated ACD partly through the inhibition of macrophage activation and the induction of autophagic flux. Taken together, our data indicated that metformin ameliorates ACD through enhanced autophagic flux to inhibit macrophage activation and provides a potential contribution to ACD treatment.
Collapse
Affiliation(s)
- Mengjie Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine
| | - Shulan Qu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine
| | - Jun Ma
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine
| | - Xiaoyu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine
| | - Yifu Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine
| |
Collapse
|
66
|
Lin CS, Chang CC, Yeh CC, Chang YC, Chen TL, Liao CC. Outcomes after surgery in patients with diabetes who used metformin: a retrospective cohort study based on a real-world database. BMJ Open Diabetes Res Care 2020; 8:8/2/e001351. [PMID: 33257420 PMCID: PMC7705543 DOI: 10.1136/bmjdrc-2020-001351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/06/2020] [Accepted: 08/15/2020] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Limited information was available regarding the perioperative outcomes in patients with and without use of metformin. This study aims to evaluate the complications and mortality after major surgery in patients with diabetes who use metformin. RESEARCH DESIGN AND METHODS Using a real-world database of Taiwan's National Health Insurance from 2008 to 2013, we conducted a matched cohort study of 91 356 patients with diabetes aged >20 years who used metformin and later underwent major surgery. Using a propensity score-matching technique adjusted for sociodemographic characteristics, medical condition, surgery type, and anesthesia type, 91 356 controls who underwent surgery but did not use metformin were selected. Logistic regression was used to calculate the ORs with 95% CIs for postoperative complications and 30-day mortality associated with metformin use. RESULTS Patients who used metformin had a lower risk of postoperative septicemia (OR 0.94, 95% CI 0.90 to 0.98), acute renal failure (OR 0.87, 95% CI 0.79 to 0.96), and 30-day mortality (OR 0.79, 95% CI 0.71 to 0.88) compared with patients who did not use metformin, in both sexes and in every age group. Metformin users who underwent surgery also had a decreased risk of postoperative intensive care unit admission (OR 0.60, 95% CI 0.59 to 0.62) and lower medical expenditures (p<0.0001) than non-use controls. CONCLUSIONS Among patients with diabetes, those who used metformin and underwent major surgery had a lower risk of complications and mortality compared with non-users. Further randomized clinical trials are needed to show direct evidence of how metformin improves perioperative outcomes.
Collapse
Affiliation(s)
- Chao-Shun Lin
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chuen-Chau Chang
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Surgery, University of Illinois, Chicago, Illinois, USA
| | - Yi-Cheng Chang
- Division of Endocrinology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ta-Liang Chen
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chien-Chang Liao
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
67
|
Metformin Ameliorates Lipopolysaccharide-Induced Depressive-Like Behaviors and Abnormal Glutamatergic Transmission. BIOLOGY 2020; 9:biology9110359. [PMID: 33114529 PMCID: PMC7692296 DOI: 10.3390/biology9110359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/28/2022]
Abstract
Simple Summary Metformin is a promising drug for diabetes and has been reported to have antidepressant effects in depression patients or patients with comorbid depression and other diseases. However, it is largely unclear how metformin ameliorates depressive-like behaviors. To this end, we injected mice with a bacterial endotoxin (lipopolysaccharide) to induce depressive-like behaviors such as increased immobility in the forced swimming test and tail suspension test. In this depression mouse model, metformin administration ameliorated depressive-like behaviors. Glutamate is a major excitatory signal for the communications between neurons in the brain. Dysfunction of glutamatergic neurotransmission is implicated in the pathogenesis of depression. Glutamatergic transmission was elevated in our depression mouse model. Metformin administration also recovered the glutamatergic transmission deficit in the model. Taken together, our results suggest metformin had antidepressant effects and can correct abnormal glutamatergic transmission in the lipopolysaccharide-induced depression mouse model. These findings provide new insights into the underlying mechanism by which metformin acts against depression. Abstract Metformin, a first-line drug for type 2 diabetes mellitus (T2DM), has been found to reduce depressive symptoms in patients with comorbid depression and other diseases. However, it is largely unclear how metformin ameliorates depressive-like behaviors. Here, we used lipopolysaccharide (LPS) to induce depressive-like behaviors in mice and found that LPS-treated mice exhibited increased immobility in the forced swimming test (FST) and tail suspension test (TST), as well as increased glutamatergic transmission. Furthermore, metformin administration in the LPS-treated mice ameliorated depressive-like behaviors and elevated glutamatergic transmission. Our results suggest that metformin has antidepressant effects and can correct abnormal glutamatergic transmission, providing an insight into the underlying mechanism by which metformin acts against depression.
Collapse
|
68
|
Lajqi T, Pöschl J, Frommhold D, Hudalla H. The Role of Microbiota in Neutrophil Regulation and Adaptation in Newborns. Front Immunol 2020; 11:568685. [PMID: 33133082 PMCID: PMC7550463 DOI: 10.3389/fimmu.2020.568685] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Newborns are highly susceptible to infections and mainly rely on innate immune functions. Reduced reactivity, delayed activation and subsequent failure to resolve inflammation however makes the neonatal immune system a very volatile line of defense. Perinatal microbiota, nutrition and different extra-uterine factors are critical elements that define long-term outcomes and shape the immune system during the neonatal period. Neutrophils are first responders and represent a vital component of the immune system in newborns. They have long been regarded as merely executive immune cells, however this notion is beginning to shift. Neutrophils are shaped by their surrounding and adaptive elements have been described. The role of “innate immune memory” and the main triangle connection microbiome—neutrophil—adaptation will be discussed in this review.
Collapse
Affiliation(s)
- Trim Lajqi
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - Johannes Pöschl
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - David Frommhold
- Klinik für Kinderheilkunde und Jugendmedizin, Memmingen, Germany
| | - Hannes Hudalla
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| |
Collapse
|
69
|
Thangaraju P, Varthya SB, Venkatesan S. Target/therapies for chronic recurrent erythema nodosum leprosum. Indian J Pharmacol 2020; 52:222-226. [PMID: 32874007 PMCID: PMC7446680 DOI: 10.4103/ijp.ijp_788_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/12/2020] [Accepted: 03/25/2020] [Indexed: 12/04/2022] Open
Abstract
A Type 2 lepra reaction or erythema nodosum leprosum is an anticipated complication in the lepromatous spectrum of leprosy cases. It is an example of an immune complex-mediated complement activated disease (Type III hypersensitivity reaction). Hence, we tried to target the inflammatory mediators and the mental stressors for the possible management strategies.
Collapse
Affiliation(s)
- Pugazhenthan Thangaraju
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Shoban Babu Varthya
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sajitha Venkatesan
- Department of Microbiology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| |
Collapse
|
70
|
Chee YJ, Tan SK, Yeoh E. Dissecting the interaction between COVID-19 and diabetes mellitus. J Diabetes Investig 2020; 11:1104-1114. [PMID: 32558211 PMCID: PMC7323255 DOI: 10.1111/jdi.13326] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic that is caused by a novel coronavirus, severe acute respiratory syndrome coronavirus-2. Data from several countries have shown higher morbidity and mortality among individuals with chronic metabolic diseases, such as diabetes mellitus. In this review, we explore the contributing factors for poorer prognosis in these individuals. As a significant proportion of patients with COVID-19 also have diabetes mellitus, this adds another layer of complexity to their management. We explore potential interactions between antidiabetic medications and renin-angiotensin-aldosterone system inhibitors with COVID-19. Suggested recommendations for the use of antidiabetic medications for COVID-19 patients with diabetes mellitus are provided. We also review pertinent clinical considerations in the management of diabetic ketoacidosis in COVID-19 patients. In addition, we aim to increase clinicians' awareness of the metabolic effects of promising drug therapies for COVID-19. Finally, we highlight the importance of timely vaccinations for patients with diabetes mellitus.
Collapse
Affiliation(s)
- Ying Jie Chee
- Division of EndocrinologyDepartment of MedicineKhoo Teck Puat HospitalSingapore
| | - Seng Kiong Tan
- Division of EndocrinologyDepartment of MedicineKhoo Teck Puat HospitalSingapore
- Diabetes CenterAdmiralty Medical CenterSingapore
| | - Ester Yeoh
- Division of EndocrinologyDepartment of MedicineKhoo Teck Puat HospitalSingapore
- Diabetes CenterAdmiralty Medical CenterSingapore
| |
Collapse
|
71
|
Chen X, Guo H, Qiu L, Zhang C, Deng Q, Leng Q. Immunomodulatory and Antiviral Activity of Metformin and Its Potential Implications in Treating Coronavirus Disease 2019 and Lung Injury. Front Immunol 2020; 11:2056. [PMID: 32973814 PMCID: PMC7461864 DOI: 10.3389/fimmu.2020.02056] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/28/2020] [Indexed: 01/08/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19), a disease which causes severe lung injury and multiple organ damage, presents an urgent need for new drugs. The case severity and fatality of COVID-19 are associated with excessive inflammation, namely, a cytokine storm. Metformin, a widely used drug to treat type 2 diabetes (T2D) mellitus and metabolic syndrome, has immunomodulatory activity that reduces the production of proinflammatory cytokines using macrophages and causes the formation of neutrophil extracellular traps (NETs). Metformin also inhibits the cytokine production of pathogenic Th1 and Th17 cells. Importantly, treatment with metformin alleviates various lung injuries in preclinical animal models. In addition, a recent proteomic study revealed that metformin has the potential to directly inhibit SARS-CoV-2 infection. Furthermore, retrospective clinical studies have revealed that metformin treatment reduces the mortality of T2D with COVID-19. Therefore, metformin has the potential to be repurposed to treat patients with COVID-19 at risk of developing severe illness. This review summarizes the immune pathogenesis of SARS-CoV-2 and addresses the effects of metformin on inhibiting cytokine storms and preventing SARS-CoV-2 infection, as well as its side effects.
Collapse
Affiliation(s)
- Xianyang Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Huifang Guo
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Li Qiu
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Chengdong Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Qiang Deng
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qibin Leng
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
72
|
He Y, Ren E, Lu Z, Chen H, Qin Z, Wang J, He M, Liu G, Zheng L, Zhao J. Rational engineering of ferritin nanocages for targeted therapy of osteoarthritis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102210. [DOI: 10.1016/j.nano.2020.102210] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 02/08/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022]
|
73
|
Astragalus membranaceus Injection Suppresses Production of Interleukin-6 by Activating Autophagy through the AMPK-mTOR Pathway in Lipopolysaccharide-Stimulated Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1364147. [PMID: 32724488 PMCID: PMC7364262 DOI: 10.1155/2020/1364147] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022]
Abstract
Astragalus membranaceus (AM), used in traditional Chinese medicine, has been shown to enhance immune functions, and recently, its anti-inflammatory effects were identified. However, the mechanisms of action remain unclear. Most studies have shown that autophagy might be involved in the immune response of the body, including inflammation. Here, we developed an inflammatory model by stimulating macrophages with lipopolysaccharides (LPS) to explore the anti-inflammatory effect and mechanisms of AM injection from the perspective of the regulation of autophagy. Immunoblot, immunofluorescence, and ELISA were used to determine the effects of AM injection on the production of interleukin-6 (IL-6) and alterations of autophagy markers. It was found that AM injection reduced the expression of IL-6 in LPS-stimulated macrophages and reversed the LPS-induced inhibition of cellular autophagy. After treatment with inhibitors of signaling pathways, it was shown that LPS downregulated autophagy and upregulated the production of IL-6 in macrophages via the protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway. AM injection reversed the effects of LPS by activating the AMP-activated protein kinase (AMPK) instead of inhibiting Akt. These results were further confirmed by testing activators and siRNA silencing of AMPK. Hence, these 2 distinct signaling molecules appear to exert opposite effects on mTOR, which integrates information from multiple upstream signaling pathways, negatively regulating autophagy. In addition, we demonstrated that autophagy might play a key role in regulating the production of IL-6 by testing activators and inhibitors of autophagy and siRNA silencing of ATG5. These findings showed that AM injection might enhance autophagy by activating AMPK and might further play a repressive effect on the LPS-stimulated expression of IL-6. This study explored the relationship between autophagy, signaling pathways, and the production of inflammatory factors in a model of endotoxin infection and treatment with AM injection.
Collapse
|
74
|
Shi Y, Batibawa JW, Maiga M, Sun B, Li Y, Duan J, Sun Z. Identification and validation of metformin protects against PM 2.5-induced macrophages cytotoxicity by targeting toll like receptor pathway. CHEMOSPHERE 2020; 251:126526. [PMID: 32443237 DOI: 10.1016/j.chemosphere.2020.126526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 06/11/2023]
Abstract
Fine particle matter (PM2.5) has been extensively reported to contribute to the pathogenesis of pulmonary diseases. Recently, metformin has been reported to attenuate PM2.5 associated respiratory and cardiovascular injury, but the underling mechanism has not been discovered. Here, we performed comprehensively bioinformatics analysis and fully validation experiment to investigate the protection role of metformin and underling mechanism with RNAseq profile in GEO database. A combination of various bioinformatics tools including edgeR, principal component analysis (PCA), K-Means clustering, Gene Set Enrichment Analysis (GSEA), GO and KEGG enrichment were performed to identify the TLRs/MyD88/NF-κB axis functional as the key signaling transduction during PM2.5 associated toxicity. PM2.5 activated TLRs/MyD88/NF-κB pathway and resulted in significantly generation of IL-6, TNF-α, mitochondrial damage, decreasing of cell viability and increased LDH activity in RAW264.7 cells. Metformin significantly attenuated the production of IL-6, mitochondrial damage, cell viability and LDH activity by limiting TLRs/MyD88/NF-κB pathway. The siRNA against AMPKα2 or negative control were transfected to RAW264.7 cells to identify whether metformin protects PM2.5-induced cytotoxicity in an AMPKα2-dependent manner. Pretreatment with metformin significantly attenuated PM2.5 induced decreasing of cell viability and increased LDH activity, as well as inhibited the TLRs/MyD88/NF-κB pathway in both siControl or siAMPKα2 cells. Taken together, our results indicate that metformin protects against PM2.5-induced mitochondrial damage and cell cytotoxicity by inhibiting TLRs/MyD88/NF-κB signaling pathway in an AMPKα2 independent manner.
Collapse
Affiliation(s)
- Yanfeng Shi
- Department of Toxicology and Sanitary Chemistry, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Josevata Werelagi Batibawa
- Department of Toxicology and Sanitary Chemistry, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Modibo Maiga
- Department of Toxicology and Sanitary Chemistry, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Baiyang Sun
- Department of Toxicology and Sanitary Chemistry, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
75
|
Lee CH, Han JH, Kim S, Lee H, Kim S, Nam DH, Cho DH, Woo CH. Metformin ameliorates bile duct ligation-induced acute hepatic injury via regulation of ER stress. BMB Rep 2020. [PMID: 31791444 PMCID: PMC7330811 DOI: 10.5483/bmbrep.2020.53.6.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Cholestasis is a condition in which the bile duct becomes narrowed or clogged by a variety of factors and bile acid is not released smoothly. Bile acid-induced liver injury is facilitated by necrotic cell death, neutrophil infiltration, and inflammation. Metformin, the first-line treatment for type 2 diabetes, is known to reduce not only blood glucose but also inflammatory responses. In this study, we investigated the effects of metformin on liver injury caused by cholestasis with bile acid-induced hepatocyte injury. Static bile acid-induced liver injury is thought to be related to endoplasmic reticulum (ER) stress, inflammatory response, and chemokine expression. Metformin treatment reduced liver injury caused by bile acid, and it suppressed ER stress, inflammation, chemokine expression, and neutrophil infiltration. Similar results were obtained in mouse primary hepatocytes exposed to bile acid. Hepatocytes treated with tauroursodeoxycholic acid, an ER stress inhibitor, showed inhibition of ER stress, as well as reduced levels of inflammation and cell death. These results suggest that metformin may protect against liver injury by suppressing ER stress and inflammation and reducing chemokine expression.
Collapse
Affiliation(s)
- Chi-Ho Lee
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Jung-Hwa Han
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Sujin Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Heejung Lee
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Suji Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Dae-Hwan Nam
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Du-Hyong Cho
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Chang-Hoon Woo
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| |
Collapse
|
76
|
Zu T, Wen J, Xu L, Li H, Mi J, Li H, Brakebusch C, Fisher DE, Wu X. Up-Regulation of Activating Transcription Factor 3 in Human Fibroblasts Inhibits Melanoma Cell Growth and Migration Through a Paracrine Pathway. Front Oncol 2020; 10:624. [PMID: 32373541 PMCID: PMC7187895 DOI: 10.3389/fonc.2020.00624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
The treatment of melanoma has remained a difficult challenge. Targeting the tumor stroma has recently attracted attention for developing novel strategies for melanoma therapy. Activating transcription factor 3 (ATF3) plays a crucial role in regulating tumorigenesis and development, but whether the expression of ATF3 in human dermal fibroblasts (HDFs) can affect melanoma development hasn't been studied. Our results show that ATF3 expression is downregulated in stromal cells of human melanoma. HDFs expressing high levels of ATF3 suppressed the growth and migration of melanoma cells in association with downregulation of different cytokines including IL-6 in vitro. In vivo, HDFs with high ATF3 expression reduced tumor formation. Adding recombinant IL-6 to melanoma cells reversed those in vitro and in vivo effects, suggesting that ATF3 expression by HDFs regulates melanoma progression through the IL-6/STAT3 pathway. More importantly, HDFs pretreated with cyclosporine A or phenformin to induce ATF3 expression inhibited melanoma cell growth in vitro and in vivo. In summary, our study reveals that ATF3 suppresses human melanoma growth and that inducing the expression of ATF3 in HDFs can inhibit melanoma growth, a new potential melanoma therapeutic approach.
Collapse
Affiliation(s)
- Tingjian Zu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Jie Wen
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Lin Xu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Department of Orthodontics, Liaocheng People's Hospital, Liaocheng, China
| | - Hui Li
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
| | - Jun Mi
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hui Li
- Department of Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Cord Brakebusch
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - David E Fisher
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Xunwei Wu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
77
|
Ku HC, Cheng CF. Master Regulator Activating Transcription Factor 3 (ATF3) in Metabolic Homeostasis and Cancer. Front Endocrinol (Lausanne) 2020; 11:556. [PMID: 32922364 PMCID: PMC7457002 DOI: 10.3389/fendo.2020.00556] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022] Open
Abstract
Activating transcription factor 3 (ATF3) is a stress-induced transcription factor that plays vital roles in modulating metabolism, immunity, and oncogenesis. ATF3 acts as a hub of the cellular adaptive-response network. Multiple extracellular signals, such as endoplasmic reticulum (ER) stress, cytokines, chemokines, and LPS, are connected to ATF3 induction. The function of ATF3 as a regulator of metabolism and immunity has recently sparked intense attention. In this review, we describe how ATF3 can act as both a transcriptional activator and a repressor. We then focus on the role of ATF3 and ATF3-regulated signals in modulating metabolism, immunity, and oncogenesis. The roles of ATF3 in glucose metabolism and adipose tissue regulation are also explored. Next, we summarize how ATF3 regulates immunity and maintains normal host defense. In addition, we elaborate on the roles of ATF3 as a regulator of prostate, breast, colon, lung, and liver cancers. Further understanding of how ATF3 regulates signaling pathways involved in glucose metabolism, adipocyte metabolism, immuno-responsiveness, and oncogenesis in various cancers, including prostate, breast, colon, lung, and liver cancers, is then provided. Finally, we demonstrate that ATF3 acts as a master regulator of metabolic homeostasis and, therefore, may be an appealing target for the treatment of metabolic dyshomeostasis, immune disorders, and various cancers.
Collapse
Affiliation(s)
- Hui-Chen Ku
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Tzu Chi University, Hualien, Taiwan
- *Correspondence: Ching-Feng Cheng
| |
Collapse
|
78
|
Luo X, Hu R, Zheng Y, Liu S, Zhou Z. Metformin shows anti-inflammatory effects in murine macrophages through Dicer/microribonucleic acid-34a-5p and microribonucleic acid-125b-5p. J Diabetes Investig 2020; 11:101-109. [PMID: 31102492 PMCID: PMC6944836 DOI: 10.1111/jdi.13074] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/26/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
AIMS/INTRODUCTION Metformin, a widely prescribed antidiabetic agent, has been shown to exhibit anti-inflammatory effects in obese and type 2 diabetes patients, but the mechanism is not well elucidated. Microribonucleic acids (miRNAs) are a group of small non-coding ribonucleic acids that participate in many biological and pathological processes. The aim of the present study was to investigate whether Dicer, a key miRNA biogenesis enzyme, and miRNAs in macrophages are implicated in the anti-inflammatory effects of metformin. MATERIALS AND METHODS Enzyme-linked immunosorbent assay and reverse transcription quantitative polymerase chain reaction were carried out to verify the anti-inflammatory effects of metformin. miRNA microarray was applied to detect the expression profile of miRNA. Western-blotting, enzyme-linked immunosorbent assay and reverse transcription quantitative polymerase chain reaction were used to examine the role Dicer and miRNAs play in the anti-inflammatory effects of metformin. RESULTS In parallel with the suppression of interleukin-6 and tumor necrosis factor-α production in resting and lipopolysaccharide-stimulated macrophages, metformin could induce an increase in Dicer and most miRNAs. When Dicer was knocked down, the anti-inflammatory effects of metformin were significantly attenuated. Additionally, the upregulation of miRNA (miR)-34a-5p and miR-125b-5p by metformin were also blunted in Dicer knockdown macrophages. Furthermore, inhibition of miR-34a-5p and miR-125b-5p could impair the suppressive action of metformin on pro-inflammatory factors production, whereas overexpression of the two miRNAs mimicked the anti-inflammatory effects of metformin. CONCLUSIONS Metformin might show anti-inflammatory effects in macrophages through the induction of Dicer and the subsequent upregulation of miR-34a-5p and miR-125b-5p.
Collapse
Affiliation(s)
- Xi Luo
- Department of Metabolism and EndocrinologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Diabetes ImmunologyMinistry of EducationNational Clinical Research Center for Metabolic DiseasesCentral South UniversityChangshaHunanChina
| | - Rong Hu
- Department of Metabolism and EndocrinologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Diabetes ImmunologyMinistry of EducationNational Clinical Research Center for Metabolic DiseasesCentral South UniversityChangshaHunanChina
| | - Ying Zheng
- Center for Medical ResearchThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Shiping Liu
- Department of Metabolism and EndocrinologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Diabetes ImmunologyMinistry of EducationNational Clinical Research Center for Metabolic DiseasesCentral South UniversityChangshaHunanChina
| | - Zhiguang Zhou
- Department of Metabolism and EndocrinologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Diabetes ImmunologyMinistry of EducationNational Clinical Research Center for Metabolic DiseasesCentral South UniversityChangshaHunanChina
| |
Collapse
|
79
|
Gao J, Yuan J, Wang Q, Lei T, Shen X, Cui B, Zhang F, Ding W, Lu Z. Metformin protects against PM 2.5-induced lung injury and cardiac dysfunction independent of AMP-activated protein kinase α2. Redox Biol 2020; 28:101345. [PMID: 31669973 PMCID: PMC6838896 DOI: 10.1016/j.redox.2019.101345] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/30/2019] [Accepted: 10/15/2019] [Indexed: 01/01/2023] Open
Abstract
Fine particulate matter (PM2.5) airborne pollution increases the risk of respiratory and cardiovascular diseases. Although metformin is a well-known antidiabetic drug, it also confers protection against a series of diseases through the activation of AMP-activated protein kinase (AMPK). However, whether metformin affects PM2.5-induced adverse health effects has not been investigated. In this study, we exposed wild-type (WT) and AMPKα2-/- mice to PM2.5 every other day via intratracheal instillation for 4 weeks. After PM2.5 exposure, the AMPKα2-/- mice developed more severe lung injury and cardiac dysfunction than were developed in the WT mice; however the administration of metformin was effective in attenuating PM2.5-induced lung injury and cardiac dysfunction in both the WT and AMPKα2-/- mice. In the PM2.5-exposed mice, metformin treatment resulted in reduced systemic and pulmonary inflammation, preserved left ventricular ejection fraction, suppressed induction of pulmonary and myocardial fibrosis and oxidative stress, and increased levels of mitochondrial antioxidant enzymes. Moreover, pretreatment with metformin significantly attenuated PM2.5-induced cell death and oxidative stress in control and AMPKα2-depleted BEAS-2B and H9C2 cells, and was associated with preserved expression of mitochondrial antioxidant enzymes. These data support the notion that metformin protects against PM2.5-induced adverse health effects through a pathway that appears independent of AMPKα2. Our findings suggest that metformin may also be a novel drug for therapies that treat air pollution associated disease.
Collapse
Affiliation(s)
- Junling Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Juntao Yuan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiao'e Wang
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, 100048, China
| | - Tong Lei
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiyue Shen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bingqing Cui
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenjun Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhongbing Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
80
|
Ty MC, Loke P, Alberola J, Rodriguez A, Rodriguez-Cortes A. Immuno-metabolic profile of human macrophages after Leishmania and Trypanosoma cruzi infection. PLoS One 2019; 14:e0225588. [PMID: 31841511 PMCID: PMC6913957 DOI: 10.1371/journal.pone.0225588] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Macrophages can reprogram their metabolism in response to the surrounding stimuli, which affects their capacity to kill intracellular pathogens. We have investigated the metabolic and immune status of human macrophages after infection with the intracellular trypanosomatid parasites Leishmania donovani, L. amazonensis and T. cruzi and their capacity to respond to a classical polarizing stimulus (LPS and IFN-γ). We found that macrophages infected with Leishmania preferentially upregulate oxidative phosphorylation, which could be contributed by both host cell and parasite, while T. cruzi infection did not significantly increase glycolysis or oxidative phosphorylation. Leishmania and T. cruzi infect macrophages without triggering a strong inflammatory cytokine response, but infection does not prevent a potent response to LPS and IFN-γ. Infection appears to prime macrophages, since the cytokine response to activation with LPS and IFN-γ is more intense in infected macrophages compared to uninfected ones. Metabolic polarization in macrophages can influence infection and immune evasion of these parasites since preventing macrophage cytokine responses would help parasites to establish a persistent infection. However, macrophages remain responsive to classical inflammatory stimuli and could still trigger inflammatory cytokine secretion by macrophages.
Collapse
Affiliation(s)
- Maureen C. Ty
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
| | - P’ng Loke
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
| | - Jordi Alberola
- Dept Farmacologia, Toxicologia i Terapeutica, Facultat de Veterinaria, Edifici V, Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Ana Rodriguez
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
- * E-mail:
| | - Alheli Rodriguez-Cortes
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
- Dept Farmacologia, Toxicologia i Terapeutica, Facultat de Veterinaria, Edifici V, Universitat Autonoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
81
|
Brenig R, Pop OT, Triantafyllou E, Geng A, Singanayagam A, Perez-Shibayama C, Besse L, Cupovic J, Künzler P, Boldanova T, Brand S, Semela D, Duong FHT, Weston CJ, Ludewig B, Heim MH, Wendon J, Antoniades CG, Bernsmeier C. Expression of AXL receptor tyrosine kinase relates to monocyte dysfunction and severity of cirrhosis. Life Sci Alliance 2019; 3:3/1/e201900465. [PMID: 31822557 PMCID: PMC6907389 DOI: 10.26508/lsa.201900465] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/07/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Infectious complications in patients with cirrhosis frequently initiate episodes of decompensation and substantially contribute to the high mortality. Mechanisms of the underlying immuneparesis remain underexplored. TAM receptors (TYRO3/AXL/MERTK) are important inhibitors of innate immune responses. To understand the pathophysiology of immuneparesis in cirrhosis, we detailed TAM receptor expression in relation to monocyte function and disease severity prior to the onset of acute decompensation. TNF-α/IL-6 responses to lipopolysaccharide were attenuated in monocytes from patients with cirrhosis (n = 96) compared with controls (n = 27) and decreased in parallel with disease severity. Concurrently, an AXL-expressing (AXL+) monocyte population expanded. AXL+ cells (CD14+CD16highHLA-DRhigh) were characterised by attenuated TNF-α/IL-6 responses and T cell activation but enhanced efferocytosis and preserved phagocytosis of Escherichia coli Their expansion correlated with disease severity, complications, infection, and 1-yr mortality. AXL+ monocytes were generated in response to microbial products and efferocytosis in vitro. AXL kinase inhibition and down-regulation reversed attenuated monocyte inflammatory responses in cirrhosis ex vivo. AXL may thus serve as prognostic marker and deserves evaluation as immunotherapeutic target in cirrhosis.
Collapse
Affiliation(s)
- Robert Brenig
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland,Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Oltin T Pop
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland,Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Evangelos Triantafyllou
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Hepatology Department, St. Mary’s Hospital, Imperial College London, London, UK
| | - Anne Geng
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Arjuna Singanayagam
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Hepatology Department, St. Mary’s Hospital, Imperial College London, London, UK
| | - Christian Perez-Shibayama
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland,Institute of Immunobiology, Medical Research Centre, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Lenka Besse
- Laboratory of Experimental Oncology, Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Jovana Cupovic
- Institute of Immunobiology, Medical Research Centre, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Patrizia Künzler
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Tuyana Boldanova
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Stephan Brand
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - David Semela
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - François HT Duong
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Christopher J Weston
- Centre for Liver Research and National Institute for Health Research, Biomedical Research Unit, University of Birmingham, Birmingham, UK
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Centre, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Julia Wendon
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Charalambos G Antoniades
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Hepatology Department, St. Mary’s Hospital, Imperial College London, London, UK
| | - Christine Bernsmeier
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland,Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland,Correspondence:
| |
Collapse
|
82
|
Zhang J, Xu X, Zhu H, Wang Y, Hou Y, Liu Y. Dietary fish oil supplementation alters liver gene expressions to protect against LPS-induced liver injury in weanling piglets. Innate Immun 2019; 25:60-72. [PMID: 30782046 PMCID: PMC6830890 DOI: 10.1177/1753425918821420] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Here, the potential mechanisms of the protective effects of fish oil against
LPS-induced liver injury in a piglet model were investigated by using RNA
sequencing. Twenty-four piglets were used in a 2 × 2 factorial design, and the
main factors included diet (5% corn oil or 5% fish oil) and immunological
challenge (LPS or saline, on d 19). All piglets were slaughtered at 4 h after
challenge, and liver samples were collected. Fish oil improved liver morphology
and reduced TNF-α, IL-1β and IL-6 productions after LPS challenge. RNA
sequencing analysis showed fish oil had significant effect on the expressions of
genes involved in immune response during LPS-induced inflammation. Selected gene
expression changes were validated using quantitative RT-PCR. Fish oil reduced
the expressions of pro-inflammatory genes IL1R1,
IL1RAP, CEBPB and CRP,
and increased that of anti-inflammatory genes IL-18BP,
NFKBIA, IFIT1, IFIT2 and
ATF3. Moreover, fish oil restored the expressions of some
lipid metabolism-related genes, such as ACAA1,
ACACA, ACADS and ACADM,
which were only decreased in pigs fed a corn oil diet after LPS challenge. Our
RNA sequencing reveals novel gene-nutrient interactions following fish oil
supplementation and evoked inflammation, which add to the current understanding
of the benefits of n-3 polyunsaturated fatty acids against liver injury.
Collapse
Affiliation(s)
- Jing Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, China
| | - Xin Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, China
| | - Huiling Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, China
| | - Yang Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, China
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, China
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
83
|
Foretz M, Guigas B, Viollet B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat Rev Endocrinol 2019; 15:569-589. [PMID: 31439934 DOI: 10.1038/s41574-019-0242-2] [Citation(s) in RCA: 348] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
Despite its position as the first-line drug for treatment of type 2 diabetes mellitus, the mechanisms underlying the plasma glucose level-lowering effects of metformin (1,1-dimethylbiguanide) still remain incompletely understood. Metformin is thought to exert its primary antidiabetic action through the suppression of hepatic glucose production. In addition, the discovery that metformin inhibits the mitochondrial respiratory chain complex 1 has placed energy metabolism and activation of AMP-activated protein kinase (AMPK) at the centre of its proposed mechanism of action. However, the role of AMPK has been challenged and might only account for indirect changes in hepatic insulin sensitivity. Various mechanisms involving alterations in cellular energy charge, AMP-mediated inhibition of adenylate cyclase or fructose-1,6-bisphosphatase 1 and modulation of the cellular redox state through direct inhibition of mitochondrial glycerol-3-phosphate dehydrogenase have been proposed for the acute inhibition of gluconeogenesis by metformin. Emerging evidence suggests that metformin could improve obesity-induced meta-inflammation via direct and indirect effects on tissue-resident immune cells in metabolic organs (that is, adipose tissue, the gastrointestinal tract and the liver). Furthermore, the gastrointestinal tract also has a major role in metformin action through modulation of glucose-lowering hormone glucagon-like peptide 1 and the intestinal bile acid pool and alterations in gut microbiota composition.
Collapse
Affiliation(s)
- Marc Foretz
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Centre, Leiden, Netherlands
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France.
- CNRS, UMR8104, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
84
|
Abdulmalek SA, Balbaa M. Synergistic effect of nano-selenium and metformin on type 2 diabetic rat model: Diabetic complications alleviation through insulin sensitivity, oxidative mediators and inflammatory markers. PLoS One 2019; 14:e0220779. [PMID: 31442295 PMCID: PMC6707613 DOI: 10.1371/journal.pone.0220779] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVES In the present article, we explore a novel strategy of selenium nanoparticles (Se-NPs) for the treatment of type 2 diabetes mellitus (T2DM) by investigating the effect of Se-NPs alone and in combination with standard anti-diabetic drug metformin (MET) in high-fat diet/streptozotocin (HFD/STZ)-induced T2DM. METHODS HFD was supplemented daily to experimental rats for 8 weeks, followed by a single low dose injection of 35 mg/kg of STZ to induce T2DM. The synergistic effect of the different therapeutic strategies on diabetic complications was evaluated after the Se-NPs and MET administration for 8 weeks. Molecular and biochemical analyses were conducted to figure out the effectiveness of our treatment on insulin sensitivity, oxidative mediators and inflammatory markers. RESULTS Our observations demonstrated that HFD/STZ-induced rats have a toxic effect on serum and hepatic tissues resulted in inducing remarkable oxidative damage and hyper-inflammation with a significant disturbance in the insulin signaling pathway. Experimental animals either treated with mono-therapeutic-two doses Se-NPs (0.1 and 0.4 mg/kg) and/or MET (100 mg/kg) alone as well as the combined therapy resulted in a remarkable protective anti-diabetic effect illustrated by significant decreases in fasting blood glucose and insulin levels after 8 weeks treatment. At the same time, the levels of active insulin signaling proteins pIRS1/pAKT/pGSK-3β/pAMPK were significantly improved. Moreover, Se-NPs exhibited an anti-inflammatory effect by the mitigation of cytokine expression and a balance between oxidative stress and antioxidant status was restored. Furthermore, the anti-diabetic drug MET administration also exhibited a significant improvement in diabetic complications after the treatment period. CONCLUSION This study provides mightily the mechanism of action of combined Se-NPs and MET as a promising therapeutic alternative that synergistically alleviates most of diabetic complications and insulin resistance.
Collapse
Affiliation(s)
- Shaymaa A. Abdulmalek
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mahmoud Balbaa
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
85
|
Syu SH, Lin YW, Lin KH, Lee LM, Hsiao CH, Wen YC. Risk factors for complications and graft failure in kidney transplant patients with sepsis. Bosn J Basic Med Sci 2019; 19:304-311. [PMID: 30242808 DOI: 10.17305/bjbms.2018.3874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 09/16/2018] [Indexed: 11/16/2022] Open
Abstract
Immunosuppressive therapies decrease the incidence of acute kidney rejection after kidney transplantation, but also increase the risk of infections and sepsis. This study aimed to identify the risk factors associated with complications and/or graft failure in kidney transplant patients with sepsis. A total of 14,658 kidney transplant patients with sepsis, identified in the National Inpatient Sample (NIS) database (data from 2005-2014), were included in the study and classified into three groups: patients without complications or graft failure/dialysis (Group 1), patients with complications only (Group 2), and patients with complications and graft failure/dialysis (Group 3). Multinomial logistic regression analyses were conducted to evaluate factors associated with kidney transplant recipients. Multivariate analysis showed that, compared to Group 1, patients from Group 2 or Group 3 were more likely to be Black and to have cytomegalovirus infection, coagulopathy, and glomerulonephritis (p ≤ 0.041). Also, Group 2 was more likely to have herpes simplex virus infection, and Group 3 was more likely to have hepatitis C infection and peripheral vascular disorders, compared to Group 1 (p ≤ 0.002). In addition, patients in Group 3 were more likely to be Black and to have hepatitis C infection, peripheral vascular disorders, coagulopathy, and hypertension compared to Group 2 (p ≤ 0.039). Age and female gender were associated with lower odds of complications after kidney transplantation regardless of graft rejection/dialysis (p ≤ 0.049). Hyperlipidemia and diabetes decreased the chance of complications and graft failure/dialysis after kidney transplant (p < 0.001). In conclusion, the study highlights that black race, male gender, and specific comorbidities can increase the risk of complications and graft failure in kidney transplant patients with sepsis.
Collapse
Affiliation(s)
- Syuan-Hao Syu
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | | | | | | | | | | |
Collapse
|
86
|
Ismail Hassan F, Didari T, Khan F, Niaz K, Mojtahedzadeh M, Abdollahi M. A Review on The Protective Effects of Metformin in Sepsis-Induced Organ Failure. CELL JOURNAL 2019; 21:363-370. [PMID: 31376317 PMCID: PMC6722446 DOI: 10.22074/cellj.2020.6286] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/17/2018] [Indexed: 02/02/2023]
Abstract
Despite advances in sepsis management, it remains a major intensive-care-unit (ICU) concern. From new prospective, positive
effects of metformin, such as anti-oxidant and anti-inflammatory properties are considered potentially beneficial properties
for management of septic patients. This article reviewed the potential ameliorative effects of metformin in sepsis-induced
organ failure. Information were retrieved from PubMed, Scopus, Embase, and Google Scholar. Multi-organ damage, oxidative
stress, inflammatory cytokine stimulation, and altered circulation are hallmarks of sepsis. Metformin exerts its effect via
adenosine monophosphate-activated protein kinase (AMPK) activation. It improves sepsis-induced organ failure by inhibiting
the production of reactive oxygen species (ROS) and pro-inflammatory cytokines, preventing the activation of transcription
factors related to inflammation, decreasing neutrophil accumulation/infiltration, and also maintaining mitochondrial membrane
potential. Studies reported the safety of metformin therapeutic doses, with no evidence of lactic acidosis, in septic patients.
Collapse
Affiliation(s)
- Fatima Ismail Hassan
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Tina Didari
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Fazlullah Khan
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Kamal Niaz
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Mojtahedzadeh
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, Tehran University of Medical Sciences, Tehran, Iran.,Department of Clinical Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran. Electronic Address:.,Department of Toxicology and Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
87
|
Ma X, Jiang Z, Wang Z, Zhang Z. Administration of metformin alleviates atherosclerosis by promoting H2S production via regulating CSE expression. J Cell Physiol 2019; 235:2102-2112. [PMID: 31338841 DOI: 10.1002/jcp.29112] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/20/2019] [Indexed: 12/31/2022]
Abstract
The therapeutic effect of metformin (Met) on atherosclerosis was studied here. Effects of methionine and Met on the induction of inflammatory response and H2 S expression in peritoneal macrophages were evaluated. Enzyme-linked immunosorbent assay, immunohistochemistry assay, western blot, and quantitative reverse transcription polymerase chain reaction were conducted to observe the levels of cystathionine γ-lyase (CSE), DNA methyltransferases 1 (DNMT1), DNMT3a, DNMT3b, tumor necrosis factor (TNF- α), interleukin 1b (IL-1β), and hydrogen sulfide (H 2 S). Luciferase and bisulfite sequencing assays were also utilized to evaluate the CSE promoter activity as well as the methylation status of CSE in transfected cells. Methionine significantly elevated Hcy, TNF-a, H 2 S, and IL-1β expression while decreasing the level of CSE in C57BL/6 mice. In contrary, co-treatment with Methionine and Met reduced the detrimental effect of Methionine. Homocysteine (Hcy) decreased H 2 S expression while promoting the synthesis of IL-1β and TNF-α in THP-1 and raw264.7 cells. Treatment of THP-1 and raw264.7 cells with methionine and Met reduced the activity of methionine in dose dependently. Moreover, Hcy increased the expression of DNMT and elevated the level of methylation in the CSE promoter, whereas the co-treatment with methionine and Met attenuated the effects of Hcy. Methionine significantly decreased plasma level of CSE while increasing the severity of inflammatory responses and plasma level of Hcy, which in turn suppressed H 2 S synthesis and enhanced DNA hypermethylation of CSE promoter to promote the pathogenesis of atherosclerosis. In contrary, co-treatment with methionine and Met reduced the detrimental effect of methionine.
Collapse
Affiliation(s)
- Xiaofeng Ma
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan, University of South China, Hengyang, Hunan, China.,Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan, University of South China, Hengyang, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan, University of South China, Hengyang, Hunan, China
| | - Zhuhua Zhang
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
88
|
Pereira AF, Pereira LMS, Silva CMP, Freitas Alves BW, Barbosa JS, Pinto FMM, Pereira AC, Silva KO, Pontes RB, Alencar NMN, Lima-Júnior RCP, Vale ML. Metformin reduces c-Fos and ATF3 expression in the dorsal root ganglia and protects against oxaliplatin-induced peripheral sensory neuropathy in mice. Neurosci Lett 2019; 709:134378. [PMID: 31325582 DOI: 10.1016/j.neulet.2019.134378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022]
Abstract
Oxaliplatin is a third-generation platinum drug commonly used as the first line treatment of metastatic colorectal cancer. Oxaliplatin-based anticancer regimens course with dose-limiting neurotoxicity. The pharmacological strategies used to manage such side effect are not totally effective. Metformin is an anti-diabetic drug that is described to negatively modulate painful diabetic neuropathy. Then, this study aimed to assess the effect of metformin in the oxaliplatin-induced peripheral sensory neuropathy in mice. For that purpose, Swiss male mice were injected with oxaliplatin (1, 2 or 4 mg/kg, i.v., twice a week with a total of nine injections) alone or in combination with daily administration of metformin (250 mg/kg, p.o.). Thermal and mechanical nociceptive tests were performed once a week for five weeks. Then, the animals were euthanized on day 35 post-first injection of oxaliplatin and the dorsal root ganglia were harvested for the assessment of c-Fos and ATF3 expressions. Oxaliplatin caused a nociceptive response accompanied by the increased expression of c-Fos and ATF3 in the dorsal root ganglia and spinal cord. In addition, the oxaliplatin-associated nociception was significantly attenuated by metformin (P < 0.05), which also reduced the expression of c-Fos and ATF3 (P < 0.05). Therefore, metformin protected from the peripheral sensory neuropathy induced by oxaliplatin, which was confirmed by the reduction of c-Fos and ATF3 expression, two known neuronal activation and damage markers, respectively.
Collapse
Affiliation(s)
- Anamaria Falcão Pereira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lus Mário Silva Pereira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Bruno Wesley Freitas Alves
- Department of Morphology, Morpho-functional Sciences Post Graduation Program, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jéssica Sales Barbosa
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Ana Carolina Pereira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Karla Oliveira Silva
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Renata Bessa Pontes
- Department of Physical Therapy, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Nylane Maria Nunes Alencar
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Roberto César Pereira Lima-Júnior
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Mariana Lima Vale
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Department of Morphology, Morpho-functional Sciences Post Graduation Program, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
89
|
Nikiforov NG, Wetzker R, Kubekina MV, Petukhova AV, Kirichenko TV, Orekhov AN. Trained Circulating Monocytes in Atherosclerosis: Ex Vivo Model Approach. Front Pharmacol 2019; 10:725. [PMID: 31316385 PMCID: PMC6610245 DOI: 10.3389/fphar.2019.00725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/05/2019] [Indexed: 11/25/2022] Open
Abstract
Inflammation is one of the key processes in the pathogenesis of atherosclerosis. Numerous studies are focused on the local inflammatory processes associated with atherosclerotic plaque initiation and progression. However, changes in the activation state of circulating monocytes, the main components of the innate immunity, may precede the local events. In this article, we discuss tolerance, which results in decreased ability of monocytes to be activated by pathogens and other stimuli, and training, the ability of monocyte to potentiate the response to pathological stimuli, and their relation to atherosclerosis. We also present previously unpublished results of the experiments that our group performed with monocytes/macrophages isolated from atherosclerosis patients. Our data allow assuming the existence of relationship between the formation of monocyte training and the degree of atherosclerosis progression. The suppression of trained immunity ex vivo seems to be a perspective model for searching anti-atherogenic drugs.
Collapse
Affiliation(s)
- Nikita G Nikiforov
- National Medical Research Center of Cardiology, Institute of Experimental Cardiology, Moscow, Russia.,Institute of Gene Biology, Centre of Collective Usage, Moscow, Russia.,Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Marina V Kubekina
- Institute of Gene Biology, Centre of Collective Usage, Moscow, Russia
| | - Anna V Petukhova
- Institute of Gene Biology, Centre of Collective Usage, Moscow, Russia
| | - Tatiana V Kirichenko
- National Medical Research Center of Cardiology, Institute of Experimental Cardiology, Moscow, Russia.,Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Moscow, Russia.,Institute of Human Morphology, Moscow, Russia
| |
Collapse
|
90
|
Kurelac I, Umesh Ganesh N, Iorio M, Porcelli AM, Gasparre G. The multifaceted effects of metformin on tumor microenvironment. Semin Cell Dev Biol 2019; 98:90-97. [PMID: 31091466 DOI: 10.1016/j.semcdb.2019.05.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
The efficacy of metformin in treating cancer has been extensively investigated since epidemiologic studies associated this anti-diabetic drug with a lower risk of cancer incidence. Since tumors are complex systems, in which cancer cells coexist and interact with several different types of non-malignant cells, it is not surprising that anti-cancer drugs affect not only cancer cells, but also the abundance and functions of cells of the tumor microenvironment. Recent years have seen a wide collection of reports showing how metformin, as well as other complex I inhibitors, may influence cancer progression by modulating the phenotype of non-transformed cells in a tumor. In this review, we particularly focus on the effect of metformin on angiogenesis, cancer-associated fibroblasts, tumor-associated macrophages and cancer immunosuppression.
Collapse
Affiliation(s)
- Ivana Kurelac
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Nikkitha Umesh Ganesh
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Maria Iorio
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Anna Maria Porcelli
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy; Centro Interdipartimentale di Ricerca Industriale Scienze della Vita e Tecnologie per la Salute, Università di Bologna, Via Tolara di Sopra 41/E, 40064, Ozzano dell'Emilia, Italy.
| | - Giuseppe Gasparre
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy; Centro di Ricerca Biomedica Applicata (CRBA), Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| |
Collapse
|
91
|
Meziane W, Mekkaoui Z, Hai I, Kacimi K, Djilali K, Touil-Boukoffa C, Lefranc G, Fernandez A, Lamb N, Mennechet F, Aribi M. Combination of metformin with sodium selenite induces a functional phenotypic switch of human GM-CSF monocyte-derived macrophages. Int Immunopharmacol 2019; 73:212-224. [PMID: 31108386 DOI: 10.1016/j.intimp.2019.05.004] [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: 07/30/2018] [Revised: 04/03/2019] [Accepted: 05/04/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVES We evaluated the effects of metformin (Met, 1,1‑dimethylbiguanide hydrochloride) combined or not with sodium selenite (Ss, Na2SeO3) on the functional activities of LPS-activated GM-CSF monocyte-derived macrophages (GM-MDM). MATERIALS AND METHODS Human GM-MDMs from three healthy donors were treated with Met or Ss alone, or with the combination of Met and Ss, and assayed for various biological activities and cytokines expression. RESULTS Met alone and Ss alone had significantly different effects on phagocytosis and killing capacities and IL-β production, but had similar effects on the downregulation of inducible nitric oxide synthase (iNOS) activity, relative nicotinamide adenine dinucleotide reduced (NADH) dehydrogenase (Complex I), intracellular free calcium ions (ifCa2+), and on the upregulation of arginase activity. Additionally, iNOS activity-to-arginase activity ratio was downregulated in Met or Ss treated-GM-MDMs, and, conversely, upregulated in GM-MDMs treated with Met + Ss in combination, indicating that arginase activity dominates that of iNOS when the two treatments are associated. Moreover, combination of Met with Ss significantly upregulated hydrogen peroxide (H2O2) production and phagocytic capacity, but significantly downregulated the production of IL-1β, iNOS activity and killing capacity. On the contrary, we show that Met alone induced significant downregulation of phagocytic capacity and slight upregulation of killing capacity. Nevertheless, Ss seems to accentuate the effect of Met on the downregulation of NO production, as well as to reverse its effect on both phagocytic and killing capacities. On the other hand, all treatments induced a sharp decrease in relative levels of NADH dehydrogenase, and a marked decrease in the levels of ifCa2+. Finally, we found that GM-MDMs treated with Met or Ss, or Met combined with Ss exhibited different functional activation phenotypes, as indicated by the surface expression of co-stimulatory and cell activation and presentation molecules CD14, CD80, CD86 and HLA-DR. CONCLUSIONS Our results demonstrated that Met/Ss combination can play an important role in the modulation of functional activities of human LPS-activated GM-MDMs. Additionally, the overall effects of Met and the induction of "M2" GM-MDMs-associated arginase could be influenced by its combination with Ss.
Collapse
Affiliation(s)
- Warda Meziane
- Laboratory of Applied Molecular Biology and Immunology, BioMolim, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Zineb Mekkaoui
- Laboratory of Applied Molecular Biology and Immunology, BioMolim, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Ismahane Hai
- Laboratory of Applied Molecular Biology and Immunology, BioMolim, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Kamila Kacimi
- Laboratory of Applied Molecular Biology and Immunology, BioMolim, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Khuira Djilali
- Laboratory of Applied Molecular Biology and Immunology, BioMolim, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Chafia Touil-Boukoffa
- Cytokines and NO Synthases Team, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), BP 32 El-Alia Bab-Ezzouar, Algiers, Algeria
| | - Gérard Lefranc
- Institut de Génétique Humaine, UMR 9002 CNRS-Université de Montpellier, Montpellier, France
| | - Anne Fernandez
- Institut de Génétique Humaine, UMR 9002 CNRS-Université de Montpellier, Montpellier, France
| | - Ned Lamb
- Institut de Génétique Humaine, UMR 9002 CNRS-Université de Montpellier, Montpellier, France
| | - Franck Mennechet
- Institut de Génétique Moléculaire de Montpellier (IGMM) - UMR5535, CNRS et Université de Montpellier, France
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, BioMolim, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria.
| |
Collapse
|
92
|
Tsalamandris S, Antonopoulos AS, Oikonomou E, Papamikroulis GA, Vogiatzi G, Papaioannou S, Deftereos S, Tousoulis D. The Role of Inflammation in Diabetes: Current Concepts and Future Perspectives. Eur Cardiol 2019; 14:50-59. [PMID: 31131037 PMCID: PMC6523054 DOI: 10.15420/ecr.2018.33.1] [Citation(s) in RCA: 682] [Impact Index Per Article: 136.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a complex metabolic disorder affecting the glucose status of the human body. Chronic hyperglycaemia related to diabetes is associated with end organ failure. The clinical relationship between diabetes and atherosclerotic cardiovascular disease is well established. This makes therapeutic approaches that simultaneously target diabetes and atherosclerotic disease an attractive area for research. The majority of people with diabetes fall into two broad pathogenetic categories, type 1 or type 2 diabetes. The role of obesity, adipose tissue, gut microbiota and pancreatic beta cell function in diabetes are under intensive scrutiny with several clinical trials to have been completed while more are in development. The emerging role of inflammation in both type 1 and type 2 diabetes (T1D and T1D) pathophysiology and associated metabolic disorders, has generated increasing interest in targeting inflammation to improve prevention and control of the disease. After an extensive review of the possible mechanisms that drive the metabolic pattern in T1D and T2D and the inflammatory pathways that are involved, it becomes ever clearer that future research should focus on a model of combined suppression for various inflammatory response pathways.
Collapse
Affiliation(s)
- Sotirios Tsalamandris
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Alexios S Antonopoulos
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Evangelos Oikonomou
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - George-Aggelos Papamikroulis
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Georgia Vogiatzi
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Spyridon Papaioannou
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Spyros Deftereos
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Dimitris Tousoulis
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| |
Collapse
|
93
|
Park J, Joe Y, Ryter SW, Surh YJ, Chung HT. Similarities and Distinctions in the Effects of Metformin and Carbon Monoxide in Immunometabolism. Mol Cells 2019; 42:292-300. [PMID: 31091555 PMCID: PMC6530647 DOI: 10.14348/molcells.2019.0016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/14/2019] [Accepted: 04/18/2019] [Indexed: 12/31/2022] Open
Abstract
Immunometabolism, defined as the interaction of metabolic pathways with the immune system, influences the pathogenesis of metabolic diseases. Metformin and carbon monoxide (CO) are two pharmacological agents known to ameliorate metabolic disorders. There are notable similarities and differences in the reported effects of metformin and CO on immunometabolism. Metformin, an anti-diabetes drug, has positive effects on metabolism and can exert anti-inflammatory and anti-cancer effects via adenosine monophosphate-activated protein kinase (AMPK)-dependent and AMPK-independent mechanisms. CO, an endogenous product of heme oxygenase-1 (HO-1), can exert anti-inflammatory and antioxidant effects at low concentration. CO can confer cytoprotection in metabolic disorders and cancer via selective activation of the protein kinase R-like endoplasmic reticulum (ER) kinase (PERK) pathway. Both metformin and CO can induce mitochondrial stress to produce a mild elevation of mitochondrial ROS (mtROS) by distinct mechanisms. Metformin inhibits complex I of the mitochondrial electron transport chain (ETC), while CO inhibits ETC complex IV. Both metformin and CO can differentially induce several protein factors, including fibroblast growth factor 21 (FGF21) and sestrin2 (SESN2), which maintain metabolic homeostasis; nuclear factor erythroid 2-related factor 2 (Nrf2), a master regulator of the antioxidant response; and REDD1, which exhibits an anticancer effect. However, metformin and CO regulate these effects via different pathways. Metformin stimulates p53- and AMPK-dependent pathways whereas CO can selectively trigger the PERK-dependent signaling pathway. Although further studies are needed to identify the mechanistic differences between metformin and CO, pharmacological application of these agents may represent useful strategies to ameliorate metabolic diseases associated with altered immunometabolism.
Collapse
Affiliation(s)
- Jeongmin Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Yeonsoo Joe
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Stefan W. Ryter
- Joan and Sanford I. Weill Department of Medicine, and Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical Center, NY 10065,
USA
| | - Young-Joon Surh
- Tumor microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08733,
Korea
| | - Hun Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| |
Collapse
|
94
|
Luo F, Das A, Chen J, Wu P, Li X, Fang Z. Metformin in patients with and without diabetes: a paradigm shift in cardiovascular disease management. Cardiovasc Diabetol 2019; 18:54. [PMID: 31029144 PMCID: PMC6486984 DOI: 10.1186/s12933-019-0860-y] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/18/2019] [Indexed: 02/06/2023] Open
Abstract
With an increasing global burden of coronary artery disease (CAD), early detection and timely management of risk factors are crucial to reduce morbidity and mortality in such patients. Diabetes mellitus (DM) is considered an independent risk factor for the development of CAD. Metformin, an anti-diabetic drug, has been shown in pre-clinical and clinical studies, to lower the cardiovascular events in the DM patients. Growing evidence suggests that metformin has a protective effect on coronary artery beyond its hypoglycemic effects. Given its global availability, route of administration and cost, metformin provides an alternate/additional therapeutic option for primary and secondary prevention of CAD in DM and non-diabetics alike. Future prospective cohort-based studies and randomized clinical trials are needed to identify 'at-risk' population who may potentially benefit from metformin.
Collapse
Affiliation(s)
- Fei Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Avash Das
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jingfei Chen
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Panyun Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Xiangping Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Zhenfei Fang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| |
Collapse
|
95
|
Bakhteyari A, Nikpour P, Mostafavi FS, Eskandari N, Matinfar M, Soleimani Asl S, Aboutorabi R. Impact of Metformin and Pioglitazone on Serum Level of Tumor Necrosis Factor-Alpha and Lipid Profiles during Implantation Window in Diabetic Rats. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2019; 13:148-153. [PMID: 31037926 PMCID: PMC6500083 DOI: 10.22074/ijfs.2019.5636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/13/2018] [Indexed: 01/22/2023]
Abstract
Background The present study was designed to evaluate serum lipid profile and tumor necrosis factor-alpha (TNF-ɑ)
level in diabetic rats at implantation time. Type 2 diabetes mellitus (T2DM) could affect various systems, including
innate immune system and it causes chronic low-grade inflammation, increasing level of TNF-ɑ. Furthermore, T2DM
is often accompanied by impaired lipid profile. Metformin and pioglitazone are used as the first and second lines of
treatment for T2DM. Materials and Methods In this experimental study, 35 adult virgin female wistar rats, weighting 175-225 g, were
randomly categorized into five groups: i. Control, ii. Sham, iii. Nicotinamide (NA)+streptozotocin (STZ) induced
T2DM, iv. Diabetic+pioglitazone (20 mg/kg/day for 28 days oral administration), and v. Diabetic+metformin (100
mg/kg/day for 28 days oral administration). At the time of implantation, TNF-ɑ level in serum of rats was measured
by ELISA kit. Glucose was measured using photometric method and lipid profiles were calculated by enzymatic
methods. Results Level of TNF-ɑ in the diabetic group was significantly higher than other groups (P<0.001). In metformin
treated group, TNF-ɑ serum level was also significantly higher than pioglitazone treated group (P<0.001). Fasting
blood sugar (FBS) and lipid profiles were significantly higher in diabetic group. Conclusion Metformin and pioglitazone have similar effects on glucose, lipid profiles and TNF-ɑ serum levels.
Among these drugs, pioglitazone has more efficient influence on TNF-α serum level, in comparison with metformin.
Collapse
Affiliation(s)
- Abbas Bakhteyari
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemah Sadat Mostafavi
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Matinfar
- Department of Internal Medicine Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sara Soleimani Asl
- Endometrium and Endometriosis Research Center, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Roshanak Aboutorabi
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.Electronic Address:
| |
Collapse
|
96
|
García G, Gutiérrez-Lara EJ, Centurión D, Granados-Soto V, Murbartián J. Fructose-Induced Insulin Resistance as a Model of Neuropathic Pain in Rats. Neuroscience 2019; 404:233-245. [DOI: 10.1016/j.neuroscience.2019.01.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/21/2022]
|
97
|
Liang H, Ding X, Li L, Wang T, Kan Q, Wang L, Sun T. Association of preadmission metformin use and mortality in patients with sepsis and diabetes mellitus: a systematic review and meta-analysis of cohort studies. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:50. [PMID: 30777119 PMCID: PMC6379943 DOI: 10.1186/s13054-019-2346-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
Background Recent studies have reported that preadmission metformin users had lower mortality than non-metformin users in patients with sepsis and diabetes mellitus; however, these results are still controversial. Therefore, we conducted a systematic review and meta-analysis of published observational cohort data to determine the association between preadmission metformin use and mortality in septic adult patients with diabetes mellitus. Methods The MEDLINE, EMBASE, and Cochrane CENTRAL databases were searched from their inception to September 30, 2018. Cohort studies that evaluated the use of metformin in septic adult patients with diabetes mellitus were included. The quality of outcomes was evaluated using the Newcastle-Ottawa Scale (NOS). The inverse variance method with random effects modelling was used to calculate the pooled odds ratios (ORs) and 95% CIs. Results Five observational cohort studies (1282 patients) that were all judged as having a low risk of bias were included. In this meta-analysis, metformin use was associated with a significantly lower mortality rate (OR, 0.59; 95% CI, 0.43–0.79, P = 0.001). Conclusions This meta-analysis indicated an association between metformin use prior to admission and lower mortality in septic adult patients with diabetes mellitus. This finding suggested that the possible effect of metformin should be evaluated in future clinical trials.
Collapse
Affiliation(s)
- Huoyan Liang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, 450052, China
| | - Xianfei Ding
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, 450052, China
| | - Lifeng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tian Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lexin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia
| | - Tongwen Sun
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, 450052, China.
| |
Collapse
|
98
|
Qing L, Fu J, Wu P, Zhou Z, Yu F, Tang J. Metformin induces the M2 macrophage polarization to accelerate the wound healing via regulating AMPK/mTOR/NLRP3 inflammasome singling pathway. Am J Transl Res 2019; 11:655-668. [PMID: 30899369 PMCID: PMC6413292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/27/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Skin wound healing is a challenging problem, especially in aging or diabetic patients, which becomes more difficult to heal, and easily results in considerable public health burden. The purpose of this study was to investigate the effects of metformin on wound healing and explore its underlying mechanism. METHODS Metformin was local topical application in rat skin defect models. Alterations in the wounded skin were observed, and angiogenesis in the wound also was analyzed by immunohistochemical staining. The markers associated with differentiation macrophage were analyzed by immunofluorescence staining. The roles of AMPK singling pathway and the relative protein of NLRP3 inflammasome in wound were also analyzed by western blotting. In addition, AMPK/mTOR/NLRP3 inflammasome signaling axis was investigated to further analyze the molecular mechanism of metformin treatment on inducing M2 macrophage polarization in vitro. RESULTS Out results showed that metformin improved wound healing and angiogenesis which was paralleled by M2 macrophage polarization. We also found that the level of relative proteins of NLRP3 inflammasome was markedly decreased after metformin treatment. Furthermore, blockage of AMPK or activation of mTOR abolished the effects of metformin treatment on depressing NLRP3 inflammasome activation, M2 polarization and improving wound healing. It suggested that the treatment effects of metformin on wound healing were through regulating AMPK/mTOR/NLRP3 inflammasome signaling axis. CONCLUSION Metformin regulated AMPK/mTOR singling pathway to inhibit NLRP3 inflammasome activation, which boosted M2 macrophage polarization to accelerate the wound healing. These findings provided new insights into the molecular mechanism of metformin therapy and its therapeutic potential in wound healing.
Collapse
Affiliation(s)
- Liming Qing
- Department of Hand and Microsurgery, Xiangya Hospital of Central South University Changsha 410008, China
| | - Jinfei Fu
- Department of Hand and Microsurgery, Xiangya Hospital of Central South University Changsha 410008, China
| | - Panfeng Wu
- Department of Hand and Microsurgery, Xiangya Hospital of Central South University Changsha 410008, China
| | - Zhengbing Zhou
- Department of Hand and Microsurgery, Xiangya Hospital of Central South University Changsha 410008, China
| | - Fang Yu
- Department of Hand and Microsurgery, Xiangya Hospital of Central South University Changsha 410008, China
| | - Juyu Tang
- Department of Hand and Microsurgery, Xiangya Hospital of Central South University Changsha 410008, China
| |
Collapse
|
99
|
Lee ES, Kwon MH, Kim HM, Kim N, Kim YM, Kim HS, Lee EY, Chung CH. Dibenzoylmethane ameliorates lipid-induced inflammation and oxidative injury in diabetic nephropathy. J Endocrinol 2019; 240:169-179. [PMID: 30475214 DOI: 10.1530/joe-18-0206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/31/2018] [Indexed: 11/08/2022]
Abstract
Dibenzoylmethane (DBM) is a beta-diketone analog of curcumin. Numerous studies have shown the beneficial effects of curcumin on diabetes, obesity and diabetic complications including diabetic nephropathy. Recently, we investigated the beneficial metabolic effects of DBM on high-fat diet-induced obesity. However, the effects and mechanisms of action of DBM in the kidney are currently unknown. To investigate the renoprotective effects of DBM in type 2 diabetes, we administered DBM (100 mg/kg) orally for 12 weeks to high-fat diet-induced diabetic model mice. We used mouse renal mesangial (MES13) and macrophage (RAW 264.7) cells to examine the mechanism of action of DBM (20 μM). After DBM treatment, the albumin-to-creatinine ratio was significantly decreased compared to that of the high-fat-diet group. Moreover, damaged renal ultra-structures and functions including increased glomerular volume, glomerular basement membrane thickness and inflammatory signals were ameliorated after DBM treatment. Stimulation of MES13 and RAW264.7 cells by palmitate or high-dose glucose with lipopolysaccharides increased inflammatory signals and macrophage migration. However, these changes were reversed by DBM treatment. In addition, DBM inhibited NADPH oxidase 2 and 4 expression and oxidative DNA damage. Collectively, these data suggested that DBM prevented diabetes-induced renal injury through its anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Eun Soo Lee
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Mi-Hye Kwon
- The East Coast Research Institute of Life Science, Gangneung-Wonju National University, Gangneung, Korea
| | - Hong Min Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Nami Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - You Mi Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyeon Soo Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Eun Young Lee
- Department of Internal Medicine and Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Korea
| | - Choon Hee Chung
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
100
|
Wang H, Wei S. Tanshinol relieves lipopolysaccharide-induced inflammatory injury of HaCaT cells via down-regulation of microRNA-122. Phytother Res 2019; 33:910-918. [PMID: 30632205 DOI: 10.1002/ptr.6283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/08/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
This study investigated the effects of tanshinol (TAN) on lipopolysaccharide (LPS)-induced human keratinocytes inflammatory injury and underlying potential molecular mechanisms. Viability and apoptosis of HaCaT cells were assessed using MTT assay and Annexin V-FITC/PI staining, respectively. Quantitative reverse transcription-polymerase chain reaction was performed to measure the expression of microRNA-122 (miR-122) in HaCaT cells. Cell transfection was conducted to up-regulate the expression of miR-122. Western blotting was used to detect the protein expression levels of key factors involved in cell apoptosis, inflammatory response, c-Jun N-terminal kinase (JNK), and nuclear factor kappa B (NF-κB) pathways. We found that LPS treatment induced HaCaT cell inflammatory injury by inhibiting cell viability, promoting cell apoptosis, and enhancing the protein expression levels of cyclooxygenase 2 and inducible nitric oxide synthase. TAN treatment relieved LPS-induced HaCaT cell inflammatory injury. Moreover, TAN treatment attenuated LPS-induced activation of JNK and NF-κB pathways in HaCaT cells. Furthermore, TAN treatment alleviated LPS-induced up-regulation of miR-122. Overexpression of miR-122 reversed the effects of TAN on LPS-induced HaCaT cell inflammatory injury and activation of JNK and NF-κB pathways. In conclusion, TAN exerted anti-inflammatory and protective effects on keratinocytes injury. TAN relieved LPS-induced inflammatory injury of human HaCaT cells via down-regulating miR-122 and then inactivating JNK and NF-κB pathways.
Collapse
Affiliation(s)
- Hui Wang
- Operating Room, Jining No.1 People's Hospital, Jining, 272011, China.,Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining, 272067, China
| | - Shujing Wei
- Operating Room, Jining No.1 People's Hospital, Jining, 272011, China
| |
Collapse
|