1
|
Leinweber B, Pilorz V, Olejniczak I, Skrum L, Begemann K, Heyde I, Stenger S, Sadik CD, Oster H. Bmal1 deficiency in neutrophils alleviates symptoms induced by high-fat diet. iScience 2025; 28:112038. [PMID: 40124497 PMCID: PMC11930374 DOI: 10.1016/j.isci.2025.112038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/24/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
Physiological processes, including metabolism and immune responses, are generated by the circadian clock, driven by clock genes. Disrupting circadian rhythms through a high-fat diet promotes obesity and inflammation. Studies show that deleting the clock gene, brain, and muscle ARNT-like 1 (Bmal1) in adipose tissue leads to overeating and weight gain. We now show that Bmal1 deletion in neutrophils protects against diet-induced obesity and reduces inflammatory macrophage infiltration into epididymal white adipose tissue (eWAT), despite increased food intake over 20 weeks of a high-fat diet. This protection is linked to enhanced energy expenditure, increased UCP1 expression in iBAT, improved insulin sensitivity, and altered expression of genes encoding chemokine receptors CXCR2, CXCR4, and the ligand Cxcl2 in eWAT. Our findings reveal a key role of Bmal1 in neutrophils in regulating high-fat diet-induced adipose inflammation and emphasize circadian regulation's importance in immuno-metabolic function.
Collapse
Affiliation(s)
- Brinja Leinweber
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Violetta Pilorz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Iwona Olejniczak
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Ludmila Skrum
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Kimberly Begemann
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Isabel Heyde
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Sarah Stenger
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Christian David Sadik
- University of Lübeck, Department of Dermatology, Allergy, and Venereology Ratzeburger Allee, 23562 Luebeck, Germany
| | - Henrik Oster
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| |
Collapse
|
2
|
Suur BE, Börgeson E. SERPINA3C as a mediator of metabolic health in offspring. Nat Metab 2025; 7:245-246. [PMID: 39891021 DOI: 10.1038/s42255-024-01209-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Affiliation(s)
- Bianca E Suur
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Emma Börgeson
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
3
|
Li Y, Li RY, Zhu JY, Chen M, Mu WJ, Luo HY, Li S, Yan LJ, Yin MT, Li X, Chen HM, Guo L. Maternal exercise prevents metabolic disorders in offspring mice through SERPINA3C. Nat Metab 2025; 7:401-420. [PMID: 39891022 DOI: 10.1038/s42255-024-01213-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
Maternal exercise can improve the metabolic health of the offspring. However, the molecular mechanisms underlying the beneficial effects of maternal exercise on the offspring remain unclear. Here, we show that maternal exercise during pregnancy alleviates high-fat diet (HFD)-induced adipose inflammation and glucose intolerance in offspring mice, accompanied by upregulation of the adipokine serine protease inhibitor A3C (SERPINA3C) both in maternal adipose tissues and the fetal circulation. Adipose SERPINA3C knockdown impairs, but its overexpression in dams mimics, maternal exercise-mediated metabolic benefits in HFD-fed offspring. Maternal SERPINA3C is transported into the fetal circulation and promotes Krüppel-like factor 4 (Klf4) gene promoter demethylation in fetal preadipocytes to increase KLF4 expression, which inhibits adipose inflammation in HFD-fed offspring mice. The SERPINA3C-cathepsin G-integrin β1 axis activates phosphatidylinositol 3-kinase signalling in preadipocytes. This promotes nuclear translocation of the p110β subunit to generate phosphatidylinositol 3,4,5-trisphosphate (PIP3) in the nucleus. O-linked β-N-acetylglucosamine (O-GlcNAc) transferase then binds to PIP3 to promote ten-eleven translocation methylcytosine dioxygenase 1 (TET1) O-GlcNAcylation, thereby enhancing TET1 activity to facilitate Klf4 gene promoter demethylation. These results provide mechanistic insights into maternal exercise-mediated improvement of offspring metabolism.
Collapse
Affiliation(s)
- Yang Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Ruo-Ying Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Wang-Jing Mu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hong-Yang Luo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Shan Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Lin-Jing Yan
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Meng-Ting Yin
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xin Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hu-Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China.
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
4
|
Li M, Hou Y, Chen Y, Sun C, Liang M, Chu X, Wen X, Yuan F, Peng C, Wang C, Xie J, Zhang J. Palmitic acid promotes miRNA release from adipocyte exosomes by activating NF-κB/ER stress. Nutr Diabetes 2024; 14:75. [PMID: 39271650 PMCID: PMC11399118 DOI: 10.1038/s41387-024-00334-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
OBJECTIVE The release of adipose tissue-derived miRNAs is increased under conditions of obesity, but the exact molecular mechanisms involved have not been elucidated. This study investigated whether obesity-induced increases in palmitic acid (PA) content could activate the NF-κB/endoplasmic reticulum stress (ER stress) pathway and promote the expression and release of exosomal miRNAs in adipocytes. METHODS Abdominal adipose tissue and serum samples were collected from normal weight individuals and people with obesity to clarify the correlation of serum PA content with NF-κB/ER stress and the release of exosomal miRNAs. NF-κB and ER stress were blocked in obese mice and in vitro cultured adipocytes to demonstrate the molecular mechanisms by which PA promotes the release of exosomal miRNAs.The morphology, particle size and distribution of the exosomes were observed via transmission electron microscopy and NTA. RESULTS Accompanied by increased serum PA levels, the NF-κB/ER stress pathway was activated in the adipose tissue of people with obesity and in high-fat diet (HFD)-induced obese mice; moreover, the levels of miRNAs in both adipose tissue and serum were increased. P-p65 (Bay11-7082) and ER stress (TUDCA) blockers significantly reduced the levels of miRNAs in abdominal adipose tissue and serum, decreased blood glucose levels, and improved glucose tolerance and insulin sensitivity in obese mice. In 3T3-L1 adipocytes, high concentrations of PA activated the NF-κB/ER stress pathway and increased the expression and release of miRNAs in exosomes. P-p65 (Bay11-7082) and ER stress (TUDCA) blockers significantly reversed the increased release exosomal miRNAs cause by PA. CONCLUSIONS Obesity-induced increases in PA content increase the expression and release of miRNAs in adipocyte exosomes by activating the NF-κB/ER stress pathway.
Collapse
Affiliation(s)
- Menghuan Li
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Yanting Hou
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Yao Chen
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Chaoyue Sun
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Maodi Liang
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Xiaolong Chu
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
- Medical College of Tarim University, Tarim Road, Alaer, Xinjiang, China
| | - Xin Wen
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Fangyuan Yuan
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Chaoling Peng
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Cuizhe Wang
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China.
| | - Jianxin Xie
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China.
| | - Jun Zhang
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China.
| |
Collapse
|
5
|
Liang Y, Zhao J, Dai T, Li X, Chen L, He Z, Guo M, Zhao J, Xu L. A review of KLF4 and inflammatory disease: Current status and future perspective. Pharmacol Res 2024; 207:107345. [PMID: 39134187 DOI: 10.1016/j.phrs.2024.107345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Inflammation is the response of the human body to injury, infection, or other abnormal states, which is involved in the development of many diseases. As a member of the Krüppel-like transcription factors (KLFs) family, KLF4 plays a crucial regulatory role in physiological and pathological processes due to its unique dual domain of transcriptional activation and inhibition. A growing body of evidence has demonstrated that KLF4 plays a pivotal role in the pathogenesis of various inflammatory disorders, including inflammatory bowel disease, osteoarthritis, renal inflammation, pneumonia, neuroinflammation, and so on. Consequently, KLF4 has emerged as a promising new therapeutic target for inflammatory diseases. This review systematically generalizes the molecular regulatory network, specific functions, and mechanisms of KLF4 to elucidate its complex roles in inflammatory diseases. An in-depth study on the biological function of KLF4 is anticipated to offer a novel research perspective and potential intervention strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Yidan Liang
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jiamin Zhao
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Tengkun Dai
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Xin Li
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Longqin Chen
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Lin Xu
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
6
|
Röszer T. MicroRNA Profile of Mouse Adipocyte-Derived Extracellular Vesicles. Cells 2024; 13:1298. [PMID: 39120327 PMCID: PMC11311276 DOI: 10.3390/cells13151298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
The post-transcriptional control of gene expression is a complex and evolving field in adipocyte biology, with the premise that the delivery of microRNA (miRNA) species to the obese adipose tissue may facilitate weight loss. Cells shed extracellular vesicles (EVs) that may deliver miRNAs as intercellular messengers. However, we know little about the miRNA profile of EVs secreted by adipocytes during postnatal development. Here, we defined the miRNA cargo of EVs secreted by mouse adipocytes in two distinct phases of development: on postnatal day 6, when adipocytes are lipolytic and thermogenic, and on postnatal day 56, when adipocytes have active lipogenesis. EVs were collected from cell culture supernatants, and their miRNA profile was defined by small RNA sequencing. The most abundant miRNA of mouse adipocyte-derived EVs was mmu-miR-148a-3p. Adipocyte EVs on postnatal day 6 were hallmarked with mmu-miR-98-5p, and some miRNAs were specific to this developmental stage, such as mmu-miR-466i-5p and 12 novel miRNAs. Adipocytes on postnatal day 56 secreted mmu-miR-365-3p, and 16 miRNAs were specific to this developmental stage. The miRNA cargo of adipocyte EVs targeted gene networks of cell proliferation, insulin signaling, interferon response, thermogenesis, and lipogenesis. We provided here a database of miRNAs secreted by developing mouse adipocytes, which may be a tool for further studies on the regulation of gene networks that control mouse adipocyte development.
Collapse
Affiliation(s)
- Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
7
|
Xiong J, Sun C, Wen X, Hou Y, Liang M, Liu J, Wei Q, Yuan F, Peng C, Chen Y, Chang Y, Wang C, Zhang J. miR-548ag promotes DPP4 expression in hepatocytes through activation of TLR(7/8)/NF-κB pathway. Int J Obes (Lond) 2024; 48:941-953. [PMID: 38424257 PMCID: PMC11217002 DOI: 10.1038/s41366-024-01504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE In our previous study, we identified a notable increase in miR-548ag content after obesity, which contributes to the progression of Type 2 diabetes Mellitus(T2DM) through the up-regulation of Dipeptidyl Peptidase-4(DPP4) expression within the liver. However, the precise molecular mechanisms underlying the upregulation of DPP4 by miR-548ag remain elusive. Mature miRNAs rich in GU sequences can activate the TLR(7/8)/NF-κB signalling pathway, which transcriptionally activates DPP4 expression. Notably, the proportion of GU sequences in hsa-miR-548ag was found to be 47.6%. The study proposes a hypothesis suggesting that miR-548ag could potentially increase DPP4 expression in hepatocytes by activating the TLR(7/8)/NF-κB signalling pathway. METHODS Male C57BL/6J mice were fed normal chow diet (NCD, n = 16) or high-fat diet (HFD, n = 16) for 12 weeks. For a duration of 6 weeks, NCD mice received intraperitoneal injections of a miR-548ag mimic, while HFD mice and db/db mice (n = 16) were administered intraperitoneal injections of a miR-548ag inhibitor. qRT-PCR and Western Blot were used to detect the expression level of miR-548ag, DPP4 and the activation of TLR(7/8)/NF-κB signalling pathway. HepG2 and L02 cells were transfected with miR-548ag mimic, miR-548ag inhibitor, TLR7/8 interfering fragment, and overexpression of miR-548ag while inhibiting TLR7/8, respectively. RESULTS (1) We observed elevated levels of miR-548ag in the serum, adipose tissue, and liver of obese mice, accompanied by an upregulation of TLR7/8, pivotal protein in the NF-κB pathway, and DPP4 expression in the liver. (2) miR-548ag promotes DPP4 expression in hepatocytes via the TLR(7/8)/NF-κB signalling pathway, resulting in a reduction in the glucose consumption capacity of hepatocytes. (3) The administration of a miR-548ag inhibitor enhanced glucose tolerance and insulin sensitivity in db/db mice. CONCLUSIONS MiR-548ag promotes the expression of DPP4 in hepatocytes by activating the TLR(7/8)/NF-κB signalling pathway. MiR-548ag may be a potential target for the treatment of T2DM.
Collapse
Affiliation(s)
- Jianyu Xiong
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Chaoyue Sun
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xin Wen
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Yanting Hou
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Maodi Liang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Jie Liu
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Qianqian Wei
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Fangyuan Yuan
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Chaoling Peng
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Yao Chen
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Yongsheng Chang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China.
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300000, China.
| | - Cuizhe Wang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China.
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China.
| | - Jun Zhang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China.
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
8
|
Wang J, Liu J, Yuan C, Yang B, Pang H, Chen K, Feng J, Deng Y, Zhang X, Li W, Wang C, Xie J, Zhang J. Palmitic acid-activated GPRs/KLF7/CCL2 pathway is involved in the crosstalk between bone marrow adipocytes and prostate cancer. BMC Cancer 2024; 24:75. [PMID: 38221626 PMCID: PMC10789002 DOI: 10.1186/s12885-024-11826-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 01/03/2024] [Indexed: 01/16/2024] Open
Abstract
BACKGROUND Obesity-induced abnormal bone marrow microenvironment is one of the important risk element for bone metastasis in prostate cancer (PCa). The present study aimed to determine whether obesity-induced elevation in palmitic acid (PA), which is the most abundant of the free fatty acids (FFAs), increased CCL2 via the GPRs/KLF7 pathway in bone marrow adipocytes (BMA) to facilitate PCa growth and metastasis. METHODS We constructed a bone-tumor bearing mouse model with obesity through high-fat diet, and observed the tumor formation ability of PCa cells. In vitro, observe the effect of PA on the expression level of CCL2 in BMA through GPRs/KLF7 signaling pathway. After co-culture of BMA and PCa cells, CCK8 assay and transwell experiment were used to detect the changes in biological behavior of PCa cells stimulated by BMA. RESULTS The BMA distribution in the bone marrow cavity of BALB/c nude mice fed with the high-fat diet (HFD) was evidently higher than that in the mice fed with the normal diet (ND). Moreover, HFD-induced obesity promoted KLF7/CCL2 expression in BMA and PCa cell growth in the bone marrow cavity of the mice. In the vitro experiment, a conditioned medium with increased CCL2 obtained from the BMA cultured with PA (CM-BMA-PA) was used for culturing the PCa cell lines, which evidently enhanced the proliferation, invasion, and migration ability. KLF7 significantly increased the CCL2 expression and secretion levels in BMA by targeting the promoter region of the CCL2 gene. In addition, GPR40/120 engaged in the PA-induced high KLF7/CCL2 levels in BMA to facilitate the malignant progression of PC-3 cells. CONCLUSIONS PA-activated GPRs/KLF7/CCL2 pathway in BMA facilitates prostate cancer growth and metastasis.
Collapse
Affiliation(s)
- Jingzhou Wang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Jie Liu
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Chenggang Yuan
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Bingqi Yang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Huai Pang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Keru Chen
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Jiale Feng
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Yuchun Deng
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Xueting Zhang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Wei Li
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Cuizhe Wang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China.
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China.
| | - Jianxin Xie
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China.
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China.
| | - Jun Zhang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China.
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
9
|
Hekim MG, Kelestemur MM, Bulmus FG, Bilgin B, Bulut F, Gokdere E, Ozdede MR, Kelestimur H, Canpolat S, Ozcan M. Asprosin, a novel glucogenic adipokine: a potential therapeutic implication in diabetes mellitus. Arch Physiol Biochem 2023; 129:1038-1044. [PMID: 33663304 DOI: 10.1080/13813455.2021.1894178] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVES We aimed to investigate the effects of asprosin on diabetes with a focus on serum glucose, irisin, ghrelin, leptin levels and hepatic levels of triglycerides (TG), cholesterol, low-density lipoprotein (LDL). METHODS Asprosin (10 µg/kg) was administered intraperitoneally four times at 3-day intervals and then blood and hepatic parameters above mentioned were investigated in control and diabetic mice. RESULTS The administration of asprosin increased blood glucose level in healthy animals (p = .05) whereas it did not change blood glucose level in diabetic animals. In addition, while asprosin decreased irisin level and increased ghrelin level, it did not change leptin level in diabetic mice. Therewithal, asprosin decreased the increasing levels in hepatic TG, cholesterol, and LDL in diabetic mice. CONCLUSIONS Our novel findings implicate that asprosin may be a target molecule in preventing the development and complications of diabetes.
Collapse
Affiliation(s)
| | | | - Funda Gulcu Bulmus
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Balikesir University, Balikesir, Turkey
| | - Batuhan Bilgin
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Ferah Bulut
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Ebru Gokdere
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | | | - Haluk Kelestimur
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Sinan Canpolat
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Mete Ozcan
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
10
|
Moradi N, Fadaei R, Roozbehkia M, Nourbakhsh M, Nourbakhsh M, Razzaghy-Azar M, Larijani B. Meteorin-like Protein and Asprosin Levels in Children and Adolescents with Obesity and Their Relationship with Insulin Resistance and Metabolic Syndrome. Lab Med 2023; 54:457-463. [PMID: 36762837 DOI: 10.1093/labmed/lmac152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE Two newly discovered adipokines, including Meteorin-like protein (Metrnl) and asprosin, have been implicated in glucose and insulin metabolism. This study aimed to investigate the associations of these adipokines with obesity in children and adolescents. METHODS This study was performed on 35 normal-weight children and 35 children with obesity. Anthropometric and biochemical parameters were determined. Serum concentrations of Metrnl, asprosin, and insulin were measured using enzyme-linked immunosorbent assay. RESULTS Metrnl level was significantly lower in obese children than normal-weight children. Additionally, Metrnl was negatively correlated with body mass index (BMI), insulin, waist-to-hip ratio, and homeostatic model assessment of insulin resistance (HOMA-IR). Our results also revealed that circulating asprosin levels were significantly increased in obese children compared to the control subjects and were positively correlated with BMI, insulin, HOMA-IR, cholesterol, and LDL-C. CONCLUSION Obesity is accompanied by significant alterations in Metrnl and asprosin and therefore these adipokines, especially Metrnl, are suggested as new promising therapeutic targets for obesity and its associated metabolic imbalances.
Collapse
Affiliation(s)
- Nariman Moradi
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Metabolic Disorders Research Center, Endocrinology and Metabolism, Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Fadaei
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Roozbehkia
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, TehranIran
| | - Mitra Nourbakhsh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mona Nourbakhsh
- Hazrat Aliasghar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Razzaghy-Azar
- Hazrat Aliasghar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Metabolic Disorders Research Center, Endocrinology and Metabolism, Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Liu JF, Su G, Chen LX, Zhou JP, Gao J, Zhang JJ, Wu QH, Chen W, Chen DY, Zhang ZC. Irisin Attenuates Apoptosis Following Ischemia-Reperfusion Injury Through Improved Mitochondria Dynamics and ROS Suppression Mediated Through the PI3K/Akt/mTOR Axis. Mol Neurobiol 2023:10.1007/s12035-023-03336-5. [PMID: 37060502 DOI: 10.1007/s12035-023-03336-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/03/2023] [Indexed: 04/16/2023]
Abstract
Irisin is a muscle-derived hormone that promotes the survival of motor neurons and enhances muscle size following injury. In this study, we investigated the beneficial effects and mechanism(s) of action of irisin in response to cerebral ischemia-reperfusion injury (CIRI). Right-middle cerebral artery occlusion (MCAO) and hypoxia/reoxygenation (H/R) models were generated in C57BL/6 J mice. Mouse neuronal cell lines (NSC-34) were used to confirm the molecular mechanisms of the protection afforded by irisin in response to CIRI. We found that irisin (250 μg/kg) improved cerebral function and reduced the cerebral infarct volume following CIRI. Irisin also protected neuronal cells against ischemia-reperfusion (I/R) induced apoptosis, assessed via TUNEL, and cleaved Caspase-3 staining. Western blotting of neuronal tissue from irisin treated I/R mice showed lower expression of pro-apoptotic Bax and caspase-9 (P < 0.001 and P < 0.01) and increased levels of the pro-survival protein Bcl-2 (P < 0.01 & P < 0.001 vs. I/R). Irisin also reduced the levels of reactive oxygen species (ROS) characterized through malondialdehyde (MDA) assays. Irisin was found to maintain mitochondrial homeostasis through the suppression of mitochondrial fission-linked dynamin-related protein 1 in CIRI mice (P < 0.01 and P < 0.05 v. I/R cohort). Moreover, mitochondrial fusion-related protein (Mfn2) and Opa1 expression were rescued following irisin treatment (P < 0.001 and P < 0.01 v. I/R cohort). Cell-based assays showed that irisin activates PI3K/AKT/mTOR signaling in the neurons of CIRI mice. Furthermore, the beneficial effects of irisin on NSC-34 cell-survival, mitochondrial function, and ROS generation were reversed by VS-5584, a highly specific PI3K/AKT/mTOR inhibitor. Collectively, these data highlight the ability of irisin to alleviate CIRI in vivo and in vitro. The mechanisms of action of irisin include the attenuation of apoptosis through the prevention of mitochondrial fission and increased mitochondrial fusion and the alleviation of oxidative stress through activation of the PI3K/AKT/mTOR axis. We therefore identify irisin as a much-needed therapeutic for CIRI.
Collapse
Affiliation(s)
- Ji-Fei Liu
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Li-Xia Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Juan-Ping Zhou
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Jia-Jia Zhang
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Qiong-Hui Wu
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - De-Yi Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Zhen-Chang Zhang
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
12
|
Ertuna GN, Sahiner ES, Yilmaz FM, Ates I. The role of irisin and asprosin level in the pathophysiology of prediabetes. Diabetes Res Clin Pract 2023; 199:110642. [PMID: 36966974 DOI: 10.1016/j.diabres.2023.110642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023]
Abstract
PURPOSE We aimed to examine whether irisin and asprosin have a role in the physiopathology of prediabetes. METHODS Hundred people were selected between the age of 18-65 years for the study population (60 prediabetes, 40 healthy). For the follow-up study, the patients with prediabetes were offered a 3-month program for lifestyle change and then reevaluated. Our research is a single-center, prospective observational study. RESULTS Among the healthy group and patients with prediabetes, irisin levels were lower and asprosin levels were higher (p < 0.001) in patients. In the follow-up part, the patients' insulin levels, HOMA index scores, and asprosin levels were decreased while irisin levels were elevated (p < 0.001). Sensitivity was 98.3% and specificity was 65% for asprosin of > 56.3 ng/mL, while they were 93.3% and 65% for irisin of ≤ 120.2 pg/mL, respectively. It was found that irisin had diagnostic performance similar to insulin and the HOMA index, while asprosin performed similarly to glucose, insulin, and the HOMA index. CONCLUSION Both irisin and asprosin have been found to be related to the prediabetes pathway and it has been shown that these molecules may be useful in daily clinical practice with diagnostic performances similar to those of the HOMA index and insulin.
Collapse
Affiliation(s)
- Gokcen Nailer Ertuna
- Health Sciences University, Ankara City Hospital, Department of Internal Medicine, Ankara, Turkey.
| | - Enes Seyda Sahiner
- Health Sciences University, Ankara City Hospital, Department of Internal Medicine, Ankara, Turkey
| | - Fatma Meric Yilmaz
- Yıldırım Beyazit University, School of Medicine, Ankara City Hospital, Department of Biochemistry, Ankara, Turkey
| | - Ihsan Ates
- Health Sciences University, Ankara City Hospital, Department of Internal Medicine, Ankara, Turkey
| |
Collapse
|
13
|
Mukherjee S, Maheshwari D, Pal R, Sachdeva N. Pancreatic fat in type 2 diabetes: Causal or coincidental? World J Meta-Anal 2023; 11:68-78. [DOI: 10.13105/wjma.v11.i3.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/27/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023] Open
Abstract
Type 2 diabetes (T2D) is a multifactorial metabolic disorder affecting more than 450 million people across the globe. With the increasing prevalence of T2D and obesity, the role of fat accumulation at sites other than subcutaneous adipose tissue has received significant attention in the pathophysiology of T2D. Over the past decade and a half, a pressing concern has emerged on investigating the association of pancreatic fat accumulation or pancreatic steatosis with the development of disease. While a few reports have suggested a possible association between pancreatic fat and T2D and/or impaired glucose metabolism, a few reports suggest a lack of such association. Pancreatic fat has also been linked with genetic risk of developing T2D, prediabetes, reduced insulin secretion, and beta cell dysfunction albeit some confounding factors such as age and ethnicity may affect the outcome. With the technological advancements in clinical imaging and progress in assessment of pancreatic beta cell function, our understanding of the role of pancreatic fat in causing insulin resistance and development of various etiologies of T2D has significantly improved. This review summarizes various findings on the possible association of pancreatic fat accumulation with the pathophysiology of T2D.
Collapse
Affiliation(s)
- Soham Mukherjee
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Deep Maheshwari
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Rimesh Pal
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
14
|
Annie-Mathew AS, Prem-Santhosh S, Jayasuriya R, Ganesh G, Ramkumar KM, Sarada DVL. The pivotal role of Nrf2 activators in adipocyte biology. Pharmacol Res 2021; 173:105853. [PMID: 34455076 DOI: 10.1016/j.phrs.2021.105853] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/07/2023]
Abstract
Adipose tissue is instrumental in maintaining metabolic homeostasis by regulating energy storage in the form of triglycerides. In the case of over-nutrition, adipocytes favorably regulate lipogenesis over lipolysis and accumulate excess triglycerides, resulting in increased adipose tissue mass. An abnormal increase in hypertrophic adipocytes is associated with chronic complications such as insulin resistance, obesity, diabetes, atherosclerosis and nonalcoholic fatty liver disease. Experimental studies indicate the occurrence of oxidative stress in the pathogenesis of obesity. A common underlying link between increasing adipose tissue mass and oxidative stress is the Nuclear Factor Erythroid 2-related factor 2 (Nrf2), Keap1-Nrf2-ARE signaling, which plays an indispensable role in metabolic homeostasis by regulating oxidative and inflammatory responses. Additionally, Nrf2 also activates CCAAT/enhancer-binding protein α, (C/EBP-α), C/EBP-β and peroxisome proliferator-activated receptor γ (PPARγ) the crucial pro-adipogenic factors that promote de novo adipogenesis. Hence, at the forefront of research is the quest for prospecting novel compounds to modulate Nrf2 activity in the context of adipogenesis and obesity. This review summarizes the molecular mechanism behind the activation of the Keap1-Nrf2-ARE signaling network and the role of Nrf2 activators in adipocyte pathophysiology.
Collapse
Affiliation(s)
- A S Annie-Mathew
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Subramanian Prem-Santhosh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Ravichandran Jayasuriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Goutham Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - D V L Sarada
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
15
|
Zhang CL, Wang JJ, Li JN, Yang Y. Nonalcoholic fatty pancreas disease: An emerging clinical challenge. World J Clin Cases 2021; 9:6624-6638. [PMID: 34447810 PMCID: PMC8362510 DOI: 10.12998/wjcc.v9.i23.6624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/20/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty pancreas disease (NAFPD) is an emerging disease that has gained an increasing amount of attention in recent years. It describes fat accumulation in the pancreas with insignificant alcohol consumption, but the pathogenesis is largely unknown. A wide range of terms have been used to describe the phenomenon of pancreatic fat accumulation, but NAFPD remains an under-recognized and non-independent disorder. Obesity, age, sex, race, and unhealthy lifestyle are established independent risk factors for NAFPD, which is strongly associated with metabolic syndrome, type 2 diabetes, pancreatitis, pancreatic fistula, pancreatic cancer, and nonalcoholic fatty liver disease. At present, imaging techniques are common diagnostic aids, but uniform criteria and consensus are lacking. Therapeutically, healthy diet, weight loss, and exercise are the mainstays to reduce pancreatic fat accumulation. It can be seen that there is a limited understanding of NAFPD at this stage and further exploration is needed. Previous studies have revealed that NAFPD may directly affect diagnosis and clinical decision-making. Therefore, exploring the pathophysiological mechanism and clinical associations of NAFPD is a major challenge for researchers and clinicians.
Collapse
Affiliation(s)
- Cheng-Lei Zhang
- Department of Clinical Laboratory, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jing-Jiao Wang
- Department of Stomatology, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jian-Ning Li
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Yang
- The Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
16
|
Ceylan Hİ, Saygın Ö. An investigation of the relationship between new fasting hormone asprosin, obesity and acute-chronic exercise: current systematic review. Arch Physiol Biochem 2021; 127:373-384. [PMID: 32427509 DOI: 10.1080/13813455.2020.1767652] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The purpose of this study was to reveal the relationship between new fasting hormone asprosin, obesity, and acute-chronic exercise. The prisma guidelines were followed in forming the methodological model of this review. The articles between 2016 and 2020 (including March) were identified by scanning Google Scholar, Pub Med, and Science Direct databases. Thirty-five articles were defined from 188 articles. Three cross-sectional, and 1 prospective cohort design studies in adults, and 3 cross-sectional studies in children were found. Three randomised-control group designed studies which examined the effect of acute exercise on serum asprosin levels in obese individuals. Asprosin may be a new therapeutic biomarker to be considered in the development, but long-term and deep-rooted researches are needed, and increasing the number of studies examining the effect of exercise on asprosin in the future might help us to identify the mechanisms underlying the decrease or increase in asprosin after exercise.
Collapse
Affiliation(s)
- Halil İbrahim Ceylan
- Faculty of Kazim Karabekir Education, Physical Education and Sports Teaching Department, Ataturk University, Erzurum, Turkey
| | - Özcan Saygın
- Faculty of Sports Sciences, Coaching Science Department, Mugla Sitki Kocman University, Muğla, Turkey
| |
Collapse
|
17
|
Ghafouri-Fard S, Asadi M, Sohrabi B, Arsang-Jang S, Mehravaran E, Taheri M, Samsami M. Down-regulation of a panel of immune-related lncRNAs in breast cancer. Pathol Res Pract 2021; 224:153534. [PMID: 34175685 DOI: 10.1016/j.prp.2021.153534] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 10/21/2022]
Abstract
Breast cancer is a common neoplasm among women. This type of cancer is among malignancies in which role of long non-coding RNAs (lncRNAs) has been extensively explored. Some recently recognized lncRNAs have been less investigated in this neoplastic condition. LncRNAs that regulate tumor immunity are among those contributing in the pathogenesis of cancer. In the present expression assay, we compared expressions of nine immune-related lncRNAs namely lnc-MICAL3-2 (AC016027.1), lnc-DDX31 (AL445645.1), LINC01063, LINC02381, ENST0000615051 (AC083809.1), AC009237.14 (lnc-TRIM43B-1), ENST0000603791, LINC1234 and AC008760.1 between breast cancer samples and their paired non-cancerous samples. Expression levels of lnc-MICAL3-2, lnc-DDX31, LINC01063, LINC02381, ENST0000615051 and lnc-TRIM43B-1 were significantly decreased in breast cancer samples compared with paired control tissues (Posterior mean difference= -2.774, -2.012, -2.012, -2.015, -0.884 and -2.872; P values= 0.019, 0.0001, 0.0001, 0.0001, 0.032 and 0.0001, respectively). Expression levels of these lncRNAs have been associated with a number of clinical characteristics of breast cancer patients. Lnc-TRIM43B-1 had the highest performance in distinguishing between tumoral and non-tumoral tissues (AUC=0.82, Sensitivity=76%, Specificity=73.24%). As these lncRNAs could differentiate tumor samples from control samples, they might be regarded as putative tissue markers for breast cancer.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Asadi
- Department of Biology, Islamic Azad University, Urmia Branch, Urmia, Iran
| | - Behnoush Sohrabi
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Shahram Arsang-Jang
- Cancer Gene therapy Research Center, Zanjan University of Medical Science, Zanjan, Iran
| | - Elham Mehravaran
- Motamed Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Majid Samsami
- Department of Surgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Bretland KA, Lin L, Bretland KM, Smith MA, Fleming SM, Dengler-Crish CM. Irisin treatment lowers levels of phosphorylated tau in the hippocampus of pre-symptomatic female but not male htau mice. Neuropathol Appl Neurobiol 2021; 47:967-978. [PMID: 33768561 PMCID: PMC9292848 DOI: 10.1111/nan.12711] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022]
Abstract
AIMS Irisin is a hormone cleaved from fibronectin type-III domain-containing protein 5 in response to exercise and may be therapeutic in Alzheimer's disease (AD). Irisin is shown to repair damage caused by midlife cardiometabolic risk factors for AD (i.e., diabetes mellitus; hypertension), prevent neural amyloid beta aggregation and reduce neuroinflammation. However, there are no investigations of irisin's effect on AD-associated tauopathy in the brain. This study begins to address this gap in knowledge. METHODS Transgenic htau mice that selectively develop age-related tauopathy were treated with recombinant irisin (100 µg/kg weekly i.p.) beginning at a pre-symptomatic age (4 months) to determine if irisin could prevent emergence of early neuropathology. One month later, mice were sacrificed to collect brain tissue and serum. Protein levels of ptau (serine 202), inflammatory cytokine tumour necrosis factor alpha (TNFα) and FNDC5 were quantified using capillary-based western blotting (Wes). RESULTS Our data show that irisin treatment significantly reduced ptau and TNFα in the hippocampus and serum of female htau mice compared to vehicle-treated controls. Irisin treatment did not alter ptau levels in male htau hippocampus and appeared to enhance both neural and systemic TNFα levels. CONCLUSIONS This study provides the first evidence that enhancing the endogenous hormone irisin may be therapeutic against emerging neuropathology in a tauopathy-selective AD model. This is important because there are currently no disease-modifying therapeutics available for AD, and few agents in development address the multiple disease targets irisin appears to-making irisin an intriguing therapeutic candidate for further investigation.
Collapse
Affiliation(s)
- Katie A Bretland
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Li Lin
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Kimberly M Bretland
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA.,Kent State University, Kent, OH, USA
| | - Matthew A Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA.,Akron Children's Hospital, Rebecca D. Considine Research Institute, Akron, OH, USA
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | |
Collapse
|
19
|
The "irisin system": From biological roles to pharmacological and nutraceutical perspectives. Life Sci 2020; 267:118954. [PMID: 33359670 DOI: 10.1016/j.lfs.2020.118954] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/30/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023]
Abstract
The scientific interest in irisin, a myokine discovered in 2012, has grown exponentially in recent years. Irisin, which is mainly produced in skeletal muscle, influences the browning process of adipose tissue and lipid and energy metabolism. Recent discoveries highlight that the potential of this hormone may have been underestimated. In the first part of this review, reports on irisin structure and molecules involved in its metabolic pathway are shown. Furthermore, data related to unclear aspects are also reported: distribution, different gene expression of its precursors in different tissues, physiological levels of circulating irisin, and pharmacokinetic and pharmacodynamic profile. The second part of this work focuses on exogenous stimuli and pharmacological agents which regulate the metabolic pathway of irisin and its serum concentration. In addition to physical exercise and exposure to low temperatures, which were early recognized as exogenous stimuli able to promote the production of this myokine, preclinical and clinical evidence demonstrates the ability of natural and synthetic molecules to interfere with this metabolic pathway. Current experimental data on irisin cannot dissolve all doubts related to this interesting molecule, but they certainly underline its potential for therapeutic purposes. Thus, identification of new pharmacological tools able to act on the irisin pathway is a challenging issue for biomedical research.
Collapse
|
20
|
A state of the art review on the novel mediator asprosin in the metabolic syndrome. Porto Biomed J 2020; 5:e108. [PMID: 33324783 PMCID: PMC7732265 DOI: 10.1097/j.pbj.0000000000000108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/08/2020] [Indexed: 01/06/2023] Open
Abstract
Metabolic syndrome is a complex and heterogeneous pathology characterized by a cluster of biochemical, clinical, and metabolic factors that came together in raising the risk of cardiovascular diseases, type 2 diabetes mellitus, and all-cause mortality. Some of these features are well defined in this syndrome like: obesity, inflammation, hypertension, insulin resistance, atherosclerotic dyslipidemias, endothelial dysfunction, and inflammation. This circuit is intermediated by a complex network of hormones, cytokines, transcription factors, and adipokines, among others. Some like leptin, adiponectin, Plasminogen activator inhibitor-1, interleukin-6, Tumor necrosis factor, and their influence on the metabolic syndrome are well described in the literature and new players are described continuously. One novel player was described in 2016 by Romere et al as a fasting-induced glycogenic protein hormone named asprosin. In order to perform a state-of-the-art, nonsystematic review of asprosin, a study of the available literature was carried out in the main database (Pubmed) and the results were studied and correlated to better understand the mechanism of action of this hormone. Asprosin is not only associated with the metabolic syndrome features like glucose and lipid metabolism, insulin resistance, obesity and inflammation but also in other pathologies metabolic syndrome related like diabetic retinopathy, polycystic ovary syndrome and anorexia nervosa. A limited number of pathways were already unveiled although much more research is needed to better understand the therapeutical potential of asprosin in the metabolic syndrome.
Collapse
|
21
|
The role of non-coding RNA on macrophage modification in tuberculosis infection. Microb Pathog 2020; 149:104592. [PMID: 33098931 DOI: 10.1016/j.micpath.2020.104592] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Tuberculosis (TB), a serious disease caused by Mycobacterium tuberculosis (Mtb), remains the world's top infectious killer. It is well-established that TB can circumvent the host's immune response for long-term survival. Macrophages serve as the major host cells for TB growth and persistence and their altered functions are critical for the response of the host defense against TB exposure (elimination, latency, reactivation, and bacillary dissemination). Noncoding RNAs are crucial posttranscriptional regulators of macrophage discrimination. Therefore, this review highlights the regulatory mechanism underlying the relationship between noncoding RNAs and macrophages in TB infection, which may facilitate the identification of potential therapeutic targets and effective diagnosis biomarkers for TB disease.
Collapse
|
22
|
Wang X, Wang Y, Antony V, Sun H, Liang G. Metabolism-Associated Molecular Patterns (MAMPs). Trends Endocrinol Metab 2020; 31:712-724. [PMID: 32807598 DOI: 10.1016/j.tem.2020.07.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/06/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
Metabolic diseases pose a tremendous health threat in both developed and developing countries. The pathophysiology of metabolic diseases is complex but has been shown to be closely associated with sterile inflammation, which is initiated by various danger molecules derived from metabolic overload, such as oxidized low-density lipoproteins (OxLDLs), free fatty acids (FFAs), glucose, advanced glycation end products (AGEs), and cholesterol. These danger signals are sensed by pattern recognition receptors (PRRs) to activate proinflammatory signaling pathways and promote the release of proinflammatory mediators, leading to chronic low-grade inflammation. Although these harmful metabolic stimuli are generally regarded as damage-associated molecular patterns (DAMPs), a more specific definition and accurate classification for these DAMPs is still missing. In this opinion, we classify the harmful metabolic stimuli that can incite inflammatory responses and tissue damage via instigating PRRs as metabolism-associated molecular patterns (MAMPs), and we summarize their roles in metaflammation-mediated metabolic diseases.
Collapse
Affiliation(s)
- Xu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Victor Antony
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhuji Biomedical Institute, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhuji, Zhejiang 311800, China.
| |
Collapse
|
23
|
Wang C, Wang J, Chen K, Pang H, Li X, Zhu J, Ma Y, Qiu T, Li W, Xie J, Zhang J. Caprylic acid (C8:0) promotes bone metastasis of prostate cancer by dysregulated adipo-osteogenic balance in bone marrow. Cancer Sci 2020; 111:3600-3612. [PMID: 32770813 PMCID: PMC7540990 DOI: 10.1111/cas.14606] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer (PCa) continues to be the most common, noncutaneous cancer in men. Bone is the most frequent site of PCa metastases, and up to 90% of patients with advanced PCa develop bone metastases. An altered bone marrow microenvironment, induced by obesity, is a significant mediator for the bone tropism of PCa. However, the specific molecular mechanisms by which obesity causes changes in the bone marrow microenvironment, leading to PCa bone metastasis, are not fully understood. Our results demonstrate that a high‐fat diet (HFD) leads to dyslipidemia and changes in bone marrow of nude mice: an increase in the area and number of adipocytes and a reduction in the area and number of osteoblasts. Moreover, a HFD promoted cyclooxygenase 2 (COX2) expression and inhibited osteoprotegerin (OPG) expression in the bone microenvironment. Additionally, the total level of free fatty acids (FFAs) and caprylic acid (C8:0) was significantly higher in PCa patients with bone metastases. In vitro, caprylic acid (C8:0) promoted bone mesenchymal stem cell (MSC)‐derived adipocytic differentiation, COX2 expression, and prostaglandin E2 (PGE2) secretion, whereas osteoblastic differentiation and OPG expression were reduced. Furthermore, caprylic acid (C8:0)‐treated adipocytes promoted the invasion and migration of PCa cells. Taken together, our findings suggest caprylic acid (C8:0) promotes bone metastasis of PCa by dysregulated adipo‐osteogenic balance of bone marrow.
Collapse
Affiliation(s)
- Cuizhe Wang
- Shihezi University School of Medicine, Xinjiang, China
| | - Jingzhou Wang
- Shihezi University School of Medicine, Xinjiang, China
| | - Keru Chen
- Shihezi University School of Medicine, Xinjiang, China
| | - Huai Pang
- Shihezi University School of Medicine, Xinjiang, China
| | - Xue Li
- Shihezi University School of Medicine, Xinjiang, China
| | - Jiaojiao Zhu
- Shihezi University School of Medicine, Xinjiang, China
| | - Yinghua Ma
- Shihezi University School of Medicine, Xinjiang, China
| | - Tongtong Qiu
- Shihezi University School of Medicine, Xinjiang, China
| | - Wei Li
- Shihezi University School of Medicine, Xinjiang, China
| | - Jianxin Xie
- Shihezi University School of Medicine, Xinjiang, China
| | - Jun Zhang
- Shihezi University School of Medicine, Xinjiang, China
| |
Collapse
|
24
|
Wang Y, Zhang H, Chen Q, Jiao F, Shi C, Pei M, Lv J, Zhang H, Wang L, Gong Z. TNF-α/HMGB1 inflammation signalling pathway regulates pyroptosis during liver failure and acute kidney injury. Cell Prolif 2020; 53:e12829. [PMID: 32419317 PMCID: PMC7309595 DOI: 10.1111/cpr.12829] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Acute kidney injury (AKI) is a common complication of acute liver failure (ALF). Pyroptosis is a necrosis type related to inflammation. This study aimed to investigate the role of TNF-α/HMGB1 pathway in pyroptosis during ALF and AKI. METHODS An ALF and AKI mouse model was generated using LPS/D-Gal, and a TNF-α inhibitor, CC-5013, was used to treat the mice. THP-1 cells were induced to differentiate into M1 macrophages, then challenged with either CC-5013 or an HMGB1 inhibitor, glycyrrhizin. pLVX-mCMVZsGreen-PGK-Puros plasmids containing TNF-α wild-type (WT), mutation A94T of TNF-α and mutation P84L of TNF-α were transfected into M1 macrophages. RESULTS Treatment with CC-5013 decreased the activation of TNF-α/HMGB1 pathway and pyroptosis in the treated mice and cells compared with the control mice and cells. CC-5013 also ameliorated liver and kidney pathological changes and improved liver and renal functions in treated mice, and the number of M1 macrophages in the liver and kidney tissues also decreased. The activation of TNF-α/HMGB1 pathway and pyroptosis increased in the M1 macrophage group compared with the normal group. Similarly, the activation of TNF-α/HMGB1 pathway and pyroptosis in the LPS + WT group also increased. By contrast, the activation of the TNF-α/HMGB1 pathway and pyroptosis decreased in the LPS + A94T and LPS + P84L groups. Moreover, glycyrrhizin inhibited pyroptosis. CONCLUSION The TNF-α/HMGB1 inflammation signalling pathway plays an important role in pyroptosis during ALF and AKI.
Collapse
Affiliation(s)
- Yao Wang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Haiyue Zhang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Qian Chen
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fangzhou Jiao
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Chunxia Shi
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Maohua Pei
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jian Lv
- Department of PharmacyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hong Zhang
- Department of PharmacyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Luwen Wang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zuojiong Gong
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
25
|
|
26
|
Alan M, Gurlek B, Yilmaz A, Aksit M, Aslanipour B, Gulhan I, Mehmet C, Taner CE. Asprosin: a novel peptide hormone related to insulin resistance in women with polycystic ovary syndrome. Gynecol Endocrinol 2019; 35:220-223. [PMID: 30325247 DOI: 10.1080/09513590.2018.1512967] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Asprosin associated with insulin resistance is a newly discovered peptide hormone. The peptide promotes hepatic glucose production. Polycystic ovary syndrome (PCOS) is a metabolic disorder. Insulin resistance plays a vital role in the pathogenesis of the disease. The aim of this study was to discover the association between insulin resistance and asprosin in women with PCOS. We recruited 78 subjects with PCOS and 78 age-matched and body mass index (BMI)-matched controls into this cross-sectional study. Circulating asprosin levels were validated using ELISA method. We also determined metabolic and hormonal parameters of the involved subjects. We found that circulating asprosin levels were elevated in women with PCOS with respect to controls. Asprosin levels showed a positive correlation with insulin resistance, BMI, and free androgen index (FAI). Moreover, subjects with the highest tertile of asprosin levels represented increased odds of having PCOS as compared to those subjects with the lowest tertile asprosin levels. Increased asprosin levels resulted to high possibility of having PCOS risk associated with insulin resistance.
Collapse
Affiliation(s)
- Murat Alan
- a Department of Obstetrics and Gynecology , Izmir Tepecik Training and Research Hospital , Izmir , Turkey
| | - Beril Gurlek
- b Department of Obstetrics and Gynecology , Recep Tayyip Erdoğan University , Rize , Turkey
| | - Alpay Yilmaz
- c Department of Obstetrics and Gynecology , Katip Celebi University , Izmir , Turkey
| | - Murat Aksit
- d Department of Biochemistry and Clinical Biochemistry , Izmir Tepecik Training and Research Hospital , Izmir , Turkey
| | - Behnaz Aslanipour
- e Department of Bioengineering, Faculty of Engineering , Ege University , Izmir , Turkey
| | - Ibrahim Gulhan
- a Department of Obstetrics and Gynecology , Izmir Tepecik Training and Research Hospital , Izmir , Turkey
| | - Calan Mehmet
- f Division of Endocrinology and Metabolism, Department of Internal Medicine , Izmir Bozyaka Training and Research Hospital , Izmir , Turkey
| | - Cuneyt Eftal Taner
- a Department of Obstetrics and Gynecology , Izmir Tepecik Training and Research Hospital , Izmir , Turkey
| |
Collapse
|