51
|
Marrano N, Biondi G, Borrelli A, Cignarelli A, Perrini S, Laviola L, Giorgino F, Natalicchio A. Irisin and Incretin Hormones: Similarities, Differences, and Implications in Type 2 Diabetes and Obesity. Biomolecules 2021; 11:286. [PMID: 33671882 PMCID: PMC7918991 DOI: 10.3390/biom11020286] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
Incretins are gut hormones that potentiate glucose-stimulated insulin secretion (GSIS) after meals. Glucagon-like peptide-1 (GLP-1) is the most investigated incretin hormone, synthesized mainly by L cells in the lower gut tract. GLP-1 promotes β-cell function and survival and exerts beneficial effects in different organs and tissues. Irisin, a myokine released in response to a high-fat diet and exercise, enhances GSIS. Similar to GLP-1, irisin augments insulin biosynthesis and promotes accrual of β-cell functional mass. In addition, irisin and GLP-1 share comparable pleiotropic effects and activate similar intracellular pathways. The insulinotropic and extra-pancreatic effects of GLP-1 are reduced in type 2 diabetes (T2D) patients but preserved at pharmacological doses. GLP-1 receptor agonists (GLP-1RAs) are therefore among the most widely used antidiabetes drugs, also considered for their cardiovascular benefits and ability to promote weight loss. Irisin levels are lower in T2D patients, and in diabetic and/or obese animal models irisin administration improves glycemic control and promotes weight loss. Interestingly, recent evidence suggests that both GLP-1 and irisin are also synthesized within the pancreatic islets, in α- and β-cells, respectively. This review aims to describe the similarities between GLP-1 and irisin and to propose a new potential axis-involving the gut, muscle, and endocrine pancreas that controls energy homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francesco Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy; (N.M.); (G.B.); (A.B.); (A.C.); (S.P.); (L.L.); (A.N.)
| | | |
Collapse
|
52
|
Lee JH, Lee HS, Lee BK, Kwon YJ, Lee JW. Relationship between Muscle Mass and Non-Alcoholic Fatty Liver Disease. BIOLOGY 2021; 10:biology10020122. [PMID: 33562473 PMCID: PMC7915258 DOI: 10.3390/biology10020122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/18/2022]
Abstract
Simple Summary Sarcopenia and non-alcoholic fatty liver disease share common pathological and physiological mechanisms that can co-occur with aging. Low skeletal muscle mass index and non-alcoholic fatty liver disease were related, regardless of abdominal obesity. Maintenance of muscle mass should be emphasized for prevention of non-alcoholic fatty liver disease. Management of fatty liver also could be an important strategy to preserve muscle mass. Abstract Although sarcopenia is known to be a risk factor for non-alcoholic fatty liver disease (NAFLD), whether NAFLD is a risk factor for the development of sarcopenia is not clear. We investigated relationships between NAFLD and low skeletal muscle mass index (LSMI) using three different datasets. Participants were classified into LSMI and normal groups. LSMI was defined as a body mass index (BMI)-adjusted appendicular skeletal muscle mass <0.789 in men and <0.512 in women or as the sex-specific lowest quintile of BMI-adjusted total skeletal muscle mass. NAFLD was determined according to NAFLD liver fat score or abdominal ultrasonography. The NAFLD groups showed a higher hazard ratios (HRs) with 95% confidence intervals (CIs) for LSMI than the normal groups (HRs = 1.21, 95% CIs = 1.05–1.40). The LSMI groups also showed a higher HRs with 95% CIs for NAFLD than normal groups (HRs = 1.56, 95% CIs = 1.38–1.78). Participants with NAFLD had consistently less skeletal muscle mass over 12 years of follow-up. In conclusion, LSMI and NAFLD showed a relationship. Maintaining muscle mass should be emphasized in the management of NAFLD.
Collapse
Affiliation(s)
- Jun-Hyuk Lee
- Department of Medicine, Graduate School of Yonsei University College of Medicine, Seoul 03722, Korea;
- Department of Family Medicine, Yonsei University College of Medicine, Yongin Severance Hospital, Yongin 16995, Korea
| | - Hye-Sun Lee
- Biostatistics Collaboration Unit, Department of Research Affairs, Yonsei University College of Medicine, Seoul 06273, Korea;
| | - Byoung-Kwon Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul 06273, Korea;
| | - Yu-Jin Kwon
- Department of Family Medicine, Yonsei University College of Medicine, Yongin Severance Hospital, Yongin 16995, Korea
- Correspondence: (Y.-J.K.); (J.-W.L.); Tel.: +82-31-5189-8777 (Y.-J.K.); +82-2-2019-3480 (J.-W.L.)
| | - Ji-Won Lee
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul 06273, Korea
- Correspondence: (Y.-J.K.); (J.-W.L.); Tel.: +82-31-5189-8777 (Y.-J.K.); +82-2-2019-3480 (J.-W.L.)
| |
Collapse
|
53
|
Uda'a B, Tinduh D, Masduchi HR, Laswati L, Wibisono S, Soenatalina S, Theodora S. Comparison of Irisin Serum Healthy Untrained Males on Treadmill Exercises by Increasing Gradually Speed Vs Inclination. FOLIA MEDICA INDONESIANA 2021. [DOI: 10.20473/fmi.v56i1.24603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aerobic exercise can improve cardiorespiratory fitness in young people. Irises released into the bloodstream function to execute regulation of energy metabolism, triggering browning white adipose, which is useful in mitochondrial biogenesis. The American College of Sport Medicine (ACSM) recommends moderate intensity cardiorespiratory fitness training with a frequency of 3-5 times a week, duration of 20-60 minutes, using a treadmill. Study subjects (n = 20) were divided randomly into 2 groups, namely the moderate intensity Treadmill training group with gradual speed improvement and gradual inclination increase. Serum slices are measured on the first and last day of exercise. There was an increasing of irisin serum in the moderate intensity Treadmill training group by increasing gradually speed after 2 weeks of exercise, both 30 minutes pre-exercise (p = 0.02) and 1 hour post exercise (p = 0.01). Medium intensity Treadmill training with 2 weeks gradual speed increase with frequency 3 times a week can increase serum slice in men, healthy untrained young men.
Collapse
|
54
|
Medhat D, El-Bana MA, El-Daly SM, Ashour MN, Elias TR, Mohamed RA, Yassen NN, Abdel-Monem MA, Hussein J. Influence of irisin on diet-induced metabolic syndrome in experimental rat model. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2021; 18:347-354. [PMID: 34187125 DOI: 10.1515/jcim-2020-0030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 06/29/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To evaluate the influence of irisin on the experimental paradigm of non-alcoholic fatty liver (NAFL) as a part of MetS cluster. METHODS Forty male albino rats were divided into four groups; normal control, standard diet + irisin, high carbohydrate and fat diet (HCHF), and HCHF + irisin. After the experimental period, levels of fasting blood sugar (FBS), insulin, lipid profile, kidney functions, salusin-alpha (Sal-α), adropin, and retinol-binding protein-4 (RBP-4) were evaluated. Peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1α) expression in skeletal muscle was evaluated by quantitative real-time PCR. Aorta, liver, pancreas, and skeletal muscle tissue samples were prepared for histopathological examination. RESULTS Rats administrated HCHF showed elevated levels of FBS, lipid profile, kidney functions, RBP-4, and downregulation of PGC-1α expression along with a decline in levels of insulin, Sal-α, and adropin while administration of irisin significantly attenuated these levels. CONCLUSIONS Irisin as based therapy could emerge as a new line of treatment against MetS and its related diseases.
Collapse
Affiliation(s)
- Dalia Medhat
- Medical Biochemistry Department, National Research Centre, Doki, Giza, Egypt
| | - Mona A El-Bana
- Medical Biochemistry Department, National Research Centre, Doki, Giza, Egypt
| | - Sherien M El-Daly
- Medical Biochemistry Department, National Research Centre, Doki, Giza, Egypt
| | - Magdi N Ashour
- Medical Biochemistry Department, National Research Centre, Doki, Giza, Egypt
| | - Tahany R Elias
- Medical Biochemistry Department, National Research Centre, Doki, Giza, Egypt
| | - Rehab A Mohamed
- Medical Biochemistry Department, National Research Centre, Doki, Giza, Egypt
| | - Noha N Yassen
- Pathology Department, Medical Division, National Research Centre, Doki, Giza, Egypt
| | | | - Jihan Hussein
- Medical Biochemistry Department, National Research Centre, Doki, Giza, Egypt
| |
Collapse
|
55
|
Pervin S, Reddy ST, Singh R. Novel Roles of Follistatin/Myostatin in Transforming Growth Factor-β Signaling and Adipose Browning: Potential for Therapeutic Intervention in Obesity Related Metabolic Disorders. Front Endocrinol (Lausanne) 2021; 12:653179. [PMID: 33897620 PMCID: PMC8062757 DOI: 10.3389/fendo.2021.653179] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a global health problem and a major risk factor for several metabolic conditions including dyslipidemia, diabetes, insulin resistance and cardiovascular diseases. Obesity develops from chronic imbalance between energy intake and energy expenditure. Stimulation of cellular energy burning process has the potential to dissipate excess calories in the form of heat via the activation of uncoupling protein-1 (UCP1) in white and brown adipose tissues. Recent studies have shown that activation of transforming growth factor-β (TGF-β) signaling pathway significantly contributes to the development of obesity, and blockade or inhibition is reported to protect from obesity by promoting white adipose browning and increasing mitochondrial biogenesis. Identification of novel compounds that activate beige/brown adipose characteristics to burn surplus calories and reduce excess storage of fat are actively sought in the fight against obesity. In this review, we present recent developments in our understanding of key modulators of TGF-β signaling pathways including follistatin (FST) and myostatin (MST) in regulating adipose browning and brown adipose mass and activity. While MST is a key ligand for TGF-β family, FST can bind and regulate biological activity of several TGF-β superfamily members including activins, bone morphogenic proteins (BMP) and inhibins. Here, we review the literature supporting the critical roles for FST, MST and other proteins in modulating TGF-β signaling to influence beige and brown adipose characteristics. We further review the potential therapeutic utility of FST for the treatment of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Shehla Pervin
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, United States
| | - Srinivasa T. Reddy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Rajan Singh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, United States
- Department of Endocrinology, Men’s Health: Aging and Metabolism, Brigham and Women’s Hospital, Boston, MA, United States
- *Correspondence: Rajan Singh,
| |
Collapse
|
56
|
Liu Y, Zhu C, Guo J, Chen Y, Meng C. The Neuroprotective Effect of Irisin in Ischemic Stroke. Front Aging Neurosci 2020; 12:588958. [PMID: 33414714 PMCID: PMC7782245 DOI: 10.3389/fnagi.2020.588958] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Irisin is a PGC-1α-dependent myokine that causes increased energy expenditure by driving the development of white adipose tissue into brown fat-like tissue. Exercise can improve irisin levels and lead to its release into the blood. In ischemic stroke, neurons are always sensitive to energy supply; after a series of pathophysiological processes, reactive oxygen species that are detrimental to cell survival via mitochondrial dysfunction are generated in large quantities. As a protein associated with exercise, irisin can alleviate brain injury in the pathogenesis of ischemic stroke. It is thought that irisin can upregulate the levels of brain-derived neurotrophic factor (BDNF), which protects nerve cells from injury during ischemic stroke. Furthermore, the release of irisin into the blood via exercise influences the mitochondrial dynamics crucial to maintaining the normal function of nerve cells. Consequently, we intended to summarize the known effects of irisin during ischemic stroke.
Collapse
Affiliation(s)
- Yaqiang Liu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chunhua Zhu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiahui Guo
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yonghong Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chaoyue Meng
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
57
|
Wu F, Li Z, Cai M, Xi Y, Xu Z, Zhang Z, Li H, Zhu W, Tian Z. Aerobic exercise alleviates oxidative stress-induced apoptosis in kidneys of myocardial infarction mice by inhibiting ALCAT1 and activating FNDC5/Irisin signaling pathway. Free Radic Biol Med 2020; 158:171-180. [PMID: 32726688 DOI: 10.1016/j.freeradbiomed.2020.06.038] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022]
Abstract
Aerobic exercise involves in ameliorating kidney injury, but the underlying mechanisms are not fully clarified. In this study, we elucidated the potential mechanisms of aerobic exercise in ameliorating kidney injury following myocardial infarction (MI). In vivo, wildtype and alcat1 knockout mice were used to establish the MI model, and subjected to six-week moderate-intensity aerobic exercise. In vitro, Normal Rat Kidney (NRK) cells treated with H2O2 and recombinant human Irisin (rhIrisin) were used for exploring potential mechanisms. Our results showed that Irisin expression was up-regulated by aerobic exercise in kidneys after MI, while ALCAT1 was reduced. In alcat1 knockout mice, we found that ALCAT1 involved in the progressions of oxidative stress and apoptosis in impaired kidney tissues of MI mice, but aerobic exercise reversed these changes. Furthermore, in vitro, we observed that Irisin inhibited both H2O2-treatment or overexpression of alcat1-induced oxidative stress and apoptosis in NRK cells, partially via AMPK-Sirt1-PGC-1α pathway. These findings reveal that aerobic exercise participates in alleviating the levels of oxidative stress and apoptosis in impaired kidney tissues following MI, partially via activating FNDC5/Irisin-AMPK-Sirt1-PGC-1α signaling pathway and inhibiting ALCAT1 expression.
Collapse
Affiliation(s)
- Fangnan Wu
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Zhuo Li
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Mengxin Cai
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Yue Xi
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Zujie Xu
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Zezhou Zhang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Hangzhuo Li
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Wanyu Zhu
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China.
| |
Collapse
|
58
|
Yamashita Y, Sakakibara H, Toda T, Ashida H. Insights into the potential benefits of black soybean ( Glycine max L.) polyphenols in lifestyle diseases. Food Funct 2020; 11:7321-7339. [PMID: 32852022 DOI: 10.1039/d0fo01092h] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Black soybean (Glycine max L.), a cultivar containing abundant polyphenols in its seed coat such as anthocyanins and flavan-3-ols, has been reported to possess various health benefits toward lifestyle diseases. In this review article, the safety evaluation of polyphenol-rich black soybean seed coat extract (BE) and absorption of BE polyphenols are summarized. Additionally, we describe the antioxidant activity of BE polyphenols and their ability to induce antioxidant enzymes. The health benefits of BE and its polyphenols, such as anti-obesity and anti-hyperglycemic activities through the activation of AMP-activated protein kinase and translocation of glucose transporter 4, respectively, are also discussed. Furthermore, we found that black soybean polyphenols were involved in the improvement of vascular function. These emerging data require further investigation in scientific studies and human trials to evaluate the prevention of lifestyle diseases using black soybean polyphenols.
Collapse
Affiliation(s)
- Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| | | | - Toshiya Toda
- Department of Innovative Food Sciences, School of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya 663-8558, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
59
|
Liu S, Gao Y, Zhang L, Yin Y, Zhang W. Rspo1/Rspo3-LGR4 signaling inhibits hepatic cholesterol synthesis through the AMPKα-SREBP2 pathway. FASEB J 2020; 34:14946-14959. [PMID: 32926477 DOI: 10.1096/fj.202001234r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
R-spondins (Rspos) are endogenous ligands of leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4). Rspos-LGR4 signaling plays important roles in embryogenesis, gastrointestinal homeostasis, and food intake. Here, we investigated the impacts of Rspos-LGR4 on hepatic cholesterol synthesis. Rspo1/3 and Lgr4 knockdown mice were used to investigate the impacts of Rspo1/3-LGR4 on hepatic cholesterol synthesis. AMPKα agonist, antagonist, and shRNA were used to explore the downstream targets of Rspos-LGR4 signaling. In our study, we reported that LGR4, Rspo1, and Rspo3 were highly expressed in hepatocytes and their expressions were sensitive to energy states. Rspo1 and Rspo3 reversed OA-induced cholesterol synthesis, accompanying with increased the phosphorylation of AMPKα Thr172, reduced SREBP2 nuclear translocation, and Srebf2 mRNA expression. Conversely, hepatic LGR4 knockdown increased hepatic cholesterol synthesis and decreased the phosphorylation of AMPKα both in vitro and in vivo. Activation or inhibition of AMPKα significantly abolished the effects of LGR4 deficiency or Rspos, respectively, on cholesterol synthesis. Knockdown of AMPKα1 or/and AMPKα2 repressed Rspos-induced inhibition on cholesterol synthesis. Our study indicates that Rspo1/Rspo3-LGR4 signaling in hepatocytes suppresses cholesterol synthesis via the AMPKα-SREBP2 pathway.
Collapse
Affiliation(s)
- Shiying Liu
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuan Gao
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liping Zhang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yue Yin
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Weizhen Zhang
- School of Basic Medical Sciences, Peking University, Beijing, China.,Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
60
|
Chen W, Wang L, You W, Shan T. Myokines mediate the cross talk between skeletal muscle and other organs. J Cell Physiol 2020; 236:2393-2412. [PMID: 32885426 DOI: 10.1002/jcp.30033] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Myokines are muscle-derived cytokines and chemokines that act extensively on organs and exert beneficial metabolic functions in the whole-body through specific signal networks. Myokines as mediators provide the conceptual basis for a whole new paradigm useful for understanding how skeletal muscle communicates with other organs. In this review, we summarize and discuss classes of myokines and their physiological functions in mediating the regulatory roles of skeletal muscle on other organs and the regulation of the whole-body energy metabolism. We review the mechanisms involved in the interaction between skeletal muscle and nonmuscle organs through myokines. Moreover, we clarify the connection between exercise, myokines and disease development, which may contribute to the understanding of a potential mechanism by which physical inactivity affects the process of metabolic diseases via myokines. Based on the current findings, myokines are important factors that mediate the effect of skeletal muscle on other organ functions and whole-body metabolism.
Collapse
Affiliation(s)
- Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| |
Collapse
|
61
|
Cai L, Wu W, Lin L, Chen Y, Gao R, Shi B, Ma B, Chen Y, Jing J. Association between plasma irisin and glucose metabolism in pregnant women is modified by dietary n-3 polyunsaturated fatty acid intake. J Diabetes Investig 2020; 11:1326-1335. [PMID: 32157816 PMCID: PMC7477509 DOI: 10.1111/jdi.13249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 02/23/2020] [Accepted: 03/04/2020] [Indexed: 12/29/2022] Open
Abstract
AIMS/INTRODUCTION The role of irisin in maternal glucose metabolism and how it would respond to dietary n-3 polyunsaturated fatty acid (n-3 PUFA) intake remains unclear. This study aimed to explore whether maternal plasma irisin is associated with glucose metabolism and whether this association is modified by dietary n-3 PUFA. MATERIALS AND METHODS A total of 932 pregnant women (20-28 weeks' gestation) aged 20-45 years were recruited. Dietary n-3 PUFA was estimated using a validated quantitative food frequency questionnaire. Plasma irisin and insulin were tested by enzyme-linked immunosorbent assay, and insulin resistance (IR) was estimated using the homeostatic model assessment (HOMA). Gestational diabetes mellitus was diagnosed with a 75-g oral glucose tolerance test. Adjusted multivariable linear regression and logistic regression were carried out to examine the associations between plasma irisin and glucose metabolism. The moderating effect of dietary n-3 PUFA intake was determined by fully multiplicative models by including the interaction term. RESULTS Maternal plasma irisin was negatively associated with HOMA-IR and oral glucose tolerance test 0 h glucose level (β -0.250, -0.067; corrected P-value for false discovery rate = 0.012, 0.018, respectively), positively associated with HOMA of insulin sensitivity (β 0.028; corrected P-value for false discovery rate = 0.012), but not associated with postprandial glucose or the risk of gestational diabetes mellitus. Furthermore, we found a moderating effect of dietary n-3 PUFA on the relationships of plasma irisin with HOMA-IR and HOMA of insulin sensitivity; these associations were strengthened with increased n-3 PUFA intake (β -0.037, 0.004; P = 0.014, 0.041, respectively). CONCLUSIONS Plasma irisin was negatively associated with HOMA-IR and fasting glucose, whereas it was positively associated with HOMA of insulin sensitivity in pregnant women. We first showed that these associations were modified by dietary n-3 PUFA intake.
Collapse
Affiliation(s)
- Li Cai
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
- Guangdong Key Laboratory of NutritionDiet and HealthGuangzhouChina
| | - Weijia Wu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
- Department of Scientific ResearchHainan Women and Children's Medical CenterHaikouChina
| | - Lizi Lin
- Department of Maternal and Child HealthSchool of Public HealthPeking University Health Science CenterBeijingChina
| | - Yajun Chen
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Rui Gao
- Shenzhen Birth Cohort Study CenterNanshan Maternity and Child Healthcare Hospital of ShenzhenShenzhenChina
| | - Bijun Shi
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Bingjie Ma
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Yuming Chen
- Department of Medical Statistics and EpidemiologySchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Jin Jing
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
62
|
Dong HN, Park SY, Le CT, Choi DH, Cho EH. Irisin Regulates the Functions of Hepatic Stellate Cells. Endocrinol Metab (Seoul) 2020; 35:647-655. [PMID: 32981307 PMCID: PMC7520590 DOI: 10.3803/enm.2020.658] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Hepatic stellate cells (HSCs) are known to play a fundamental role in the progression of liver fibrosis. Once HSCs are activated, they are involved in proliferation, migration, and contractility which are characteristics of liver fibrogenesis. Recent studies have shown that irisin, a myokine secreted during physical exercise, has a protective effect in various metabolic diseases, especially in renal fibrosis. However, whether irisin is involved in HSC activation and other processes associated with liver fibrosis has not yet been investigated. In this study, we reveal the role of irisin in HSC activation as well as in proliferation, migration, and contractile properties of HSCs in vitro. METHODS LX-2 cells, immortalized human HSCs, were treated with transforming growth factor beta 1 (TGF-β1), a core regulator of HSC fibrosis, with or without irisin, and markers of the aforementioned processes were analyzed. Further, an inflammatory response was stimulated with TGF-β1 and lipopolysaccharide (LPS) in combination with irisin and the expression of cytokines was measured. RESULTS Recombinant irisin significantly suppressed the expression of TGF-β1-stimulated fibrosis markers including alpha-smooth muscle actin and collagen type 1 alpha 1 and prevented the TGF-β1-induced proliferation, migration, and contractility of LX-2 cells. Additionally, irisin ameliorated the production of interleukin-6 (IL-6) and IL-1β induced by TGF-β1 and LPS treatments. CONCLUSION These findings suggested that irisin potently improved the progression of hepatic fibrosis by regulating HSC activation, proliferation, migration, contractility, and HSC-mediated production of inflammatory cytokine.
Collapse
Affiliation(s)
- Hanh Nguyen Dong
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - So Young Park
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Cong Thuc Le
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Dae-Hee Choi
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Eun-Hee Cho
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
63
|
Bi J, Zhang J, Ren Y, Du Z, Zhang Y, Liu C, Wang Y, Zhang L, Shi Z, Wu Z, Lv Y, Wu R. Exercise hormone irisin mitigates endothelial barrier dysfunction and microvascular leakage-related diseases. JCI Insight 2020; 5:136277. [PMID: 32516137 DOI: 10.1172/jci.insight.136277] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/03/2020] [Indexed: 01/10/2023] Open
Abstract
Increased microvascular leakage is a cardinal feature of many critical diseases. Regular exercise is associated with improved endothelial function and reduced risk of cardiovascular disease. Irisin, secreted during exercise, contributes to many health benefits of exercise. However, the effects of irisin on endothelial function and microvascular leakage remain unknown. In this study, we found that irisin remarkably strengthened endothelial junctions and barrier function via binding to integrin αVβ5 receptor in LPS-treated endothelial cells. The beneficial effect of irisin was associated with suppression of the Src-MLCK-β-catenin pathway, activation of the AMPK-Cdc42/Rac1 pathway, and improvement of mitochondrial function. In preclinical models of microvascular leakage, exogenous irisin improved pulmonary function, decreased lung edema and injury, suppressed inflammation, and increased survival. In ARDS patients, serum irisin levels were decreased and inversely correlated with disease severity and mortality. In conclusion, irisin enhances endothelial barrier function and mitigates microvascular leakage-related diseases.
Collapse
Affiliation(s)
- Jianbin Bi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering.,Department of Hepatobiliary Surgery
| | - Jia Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering.,Department of Hepatobiliary Surgery
| | - Yifan Ren
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering.,Department of Hepatobiliary Surgery
| | - Zhaoqing Du
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering.,Department of Hepatobiliary Surgery
| | | | | | - Yawen Wang
- Biobank.,Department of Laboratory Medicine, and
| | - Lin Zhang
- Department of Laboratory Medicine, and
| | - Zhihong Shi
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering.,Department of Hepatobiliary Surgery
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering
| |
Collapse
|
64
|
Exercise and dietary intervention ameliorate high-fat diet-induced NAFLD and liver aging by inducing lipophagy. Redox Biol 2020; 36:101635. [PMID: 32863214 PMCID: PMC7365984 DOI: 10.1016/j.redox.2020.101635] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/25/2020] [Accepted: 07/03/2020] [Indexed: 02/08/2023] Open
Abstract
Exercise and dietary intervention are currently available strategies to treat nonalcoholic fatty liver disease (NAFLD), while the underlying mechanism remains controversial. Emerging evidence shows that lipophagy is involved in the inhibition of the lipid droplets accumulation. However, it is still unclear if exercise and dietary intervention improve NAFLD through regulating lipophagy, and how exercise of skeletal muscle can modulate lipid metabolism in liver. Moreover, NAFLD is associated with aging, and little is known about the effect of lipid accumulation on aging process. Here in vivo and in vitro models, we found that exercise and dietary intervention reduced lipid droplets formation, decreased hepatic triglyceride in the liver induced by high-fat diet. Exercise and dietary intervention enhanced the lipophagy by activating AMPK/ULK1 and inhibiting Akt/mTOR/ULK1 pathways respectively. Furthermore, exercise stimulated FGF21 production in the muscle, followed by secretion to the circulation to promote the lipophagy in the liver via an AMPK-dependent pathway. Importantly, for the first time, we demonstrated that lipid accumulation exacerbated liver aging, which was ameliorated by exercise and dietary intervention through inducing lipophagy. Our findings suggested a new mechanism of exercise and dietary intervention to improve NAFLD through promoting lipophagy. The study also provided evidence to support that muscle exercise is beneficial to other metabolic organs such as liver. The FGF21-mediated AMPK dependent lipophagy might be a potential drug target for NAFLD and aging caused by lipid metabolic dysfunction.
Collapse
|
65
|
Gonzalez-Gil AM, Elizondo-Montemayor L. The Role of Exercise in the Interplay between Myokines, Hepatokines, Osteokines, Adipokines, and Modulation of Inflammation for Energy Substrate Redistribution and Fat Mass Loss: A Review. Nutrients 2020; 12:E1899. [PMID: 32604889 PMCID: PMC7353393 DOI: 10.3390/nu12061899] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022] Open
Abstract
Exercise is an effective strategy for preventing and treating obesity and its related cardiometabolic disorders, resulting in significant loss of body fat mass, white adipose tissue browning, redistribution of energy substrates, optimization of global energy expenditure, enhancement of hypothalamic circuits that control appetite-satiety and energy expenditure, and decreased systemic inflammation and insulin resistance. Novel exercise-inducible soluble factors, including myokines, hepatokines, and osteokines, and immune cytokines and adipokines are hypothesized to play an important role in the body's response to exercise. To our knowledge, no review has provided a comprehensive integrative overview of these novel molecular players and the mechanisms involved in the redistribution of metabolic fuel during and after exercise, the loss of weight and fat mass, and reduced inflammation. In this review, we explain the potential role of these exercise-inducible factors, namely myokines, such as irisin, IL-6, IL-15, METRNL, BAIBA, and myostatin, and hepatokines, in particular selenoprotein P, fetuin A, FGF21, ANGPTL4, and follistatin. We also describe the function of osteokines, specifically osteocalcin, and of adipokines such as leptin, adiponectin, and resistin. We also emphasize an integrative overview of the pleiotropic mechanisms, the metabolic pathways, and the inter-organ crosstalk involved in energy expenditure, fat mass loss, reduced inflammation, and healthy weight induced by exercise.
Collapse
Affiliation(s)
- Adrian M. Gonzalez-Gil
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
- Tecnologico de Monterrey, Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, San Pedro Garza Garcia P.C. 66278, Mexico
| |
Collapse
|
66
|
Roles of FGF21 and irisin in obesity-related diabetes and pancreatic diseases. JOURNAL OF PANCREATOLOGY 2020. [DOI: 10.1097/jp9.0000000000000039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
67
|
Canivet CM, Bonnafous S, Rousseau D, Leclere PS, Lacas-Gervais S, Patouraux S, Sans A, Luci C, Bailly-Maitre B, Iannelli A, Tran A, Anty R, Gual P. Hepatic FNDC5 is a potential local protective factor against Non-Alcoholic Fatty Liver. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165705. [PMID: 32001301 DOI: 10.1016/j.bbadis.2020.165705] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 01/09/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
The proteolytic cleavage of Fibronectin type III domain-containing 5 (FNDC5) generates soluble irisin. Initially described as being mainly produced in muscle during physical exercise, irisin mediates adipose tissue thermogenesis and also regulates carbohydrate and lipid metabolism. The aim of this study was to evaluate the hepatic expression of FNDC5 and its role in hepatocytes in Non-Alcoholic Fatty Liver (NAFL). Here we report that hepatic expression of FNDC5 increased with hepatic steatosis and liver injury without impacting the systemic level of irisin in mouse models of NAFLD (HFD and MCDD) and in obese patients. The increased Fndc5 expression in fatty liver resulted from its upregulation in hepatocytes and non-parenchymal cells in mice. The local production of Fndc5 in hepatocytes was influenced by genotoxic stress and p53-dependent pathways. The down-regulation of FNDC5 in human HepG2 cells and in primary mouse hepatocytes increased the expression of PEPCK, a key enzyme involved in gluconeogenesis associated with a decrease in the expression of master genes involved in the VLDL synthesis (CIDEB and APOB). These alterations in FNDC5-silenced cells resulted to increased steatosis and insulin resistance in response to oleic acid and N-acetyl glucosamine, respectively. The downregulation of Fndc5 also sensitized primary hepatocytes to apoptosis in response to TNFα, which has been associated with decreased hepatoprotective autophagic flux. In conclusion, our human and experimental data strongly suggest that the hepatic expression of FNDC5 increased with hepatic steatosis and its upregulation in hepatocytes could dampen the development of NAFLD by negatively regulating steatogenesis and hepatocyte death.
Collapse
Affiliation(s)
- Clémence M Canivet
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France
| | - Stéphanie Bonnafous
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France
| | | | | | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée (CCMA), Parc Valrose, Nice, France
| | - Stéphanie Patouraux
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France
| | - Arnaud Sans
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France
| | - Carmelo Luci
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France
| | | | - Antonio Iannelli
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France
| | - Albert Tran
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France
| | - Rodolphe Anty
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France.
| | - Philippe Gual
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France.
| |
Collapse
|
68
|
Sun W, Liu P, Wang T, Wang X, Zheng W, Li J. Baicalein reduces hepatic fat accumulation by activating AMPK in oleic acid-induced HepG2 cells and high-fat diet-induced non-insulin-resistant mice. Food Funct 2020; 11:711-721. [DOI: 10.1039/c9fo02237f] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common liver disease worldwide; thus, a dietary supplement that can restrict hepatic fat accumulation is needed.
Collapse
Affiliation(s)
- Wenlong Sun
- Institute of Biomedical Research
- School of Life Sciences
- Shandong University of Technology
- Zibo
- People's Republic of China
| | - Panpan Liu
- Institute of Biomedical Research
- School of Life Sciences
- Shandong University of Technology
- Zibo
- People's Republic of China
| | - Tianqi Wang
- College of Life Science
- Yangtze University
- Jingzhou
- People's Republic of China
| | - Xudong Wang
- College of Pharmaceutical Science
- Zhejiang University of Technology
- Hangzhou
- People's Republic of China
| | - Weilong Zheng
- Institute of Biomass Resources
- Taizhou University
- Taizhou
- People's Republic of China
| | - Jingda Li
- College of Life Science
- Yangtze University
- Jingzhou
- People's Republic of China
| |
Collapse
|
69
|
Zhang C, Zhang H, Zhang M, Lin C, Wang H, Yao J, Wei Q, Lu Y, Chen Z, Xing G, Cao X. OSBPL2 deficiency upregulate SQLE expression increasing intracellular cholesterol and cholesteryl ester by AMPK/SP1 and SREBF2 signalling pathway. Exp Cell Res 2019; 383:111512. [PMID: 31356817 DOI: 10.1016/j.yexcr.2019.111512] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
Previous studies have shown that oxysterol binding protein like 2 (OSBPL2) knockdown is closely related to cholesterol metabolism. However, whether there is a direct relation between OSBPL2 and cholesterol synthesis is unknown. This study explored the mechanism of OSBPL2 deficiency in the upregulation of squalene epoxidase (SQLE) and the subsequent accumulation of intracellular cholesterol and cholesteryl ester. Here, we constructed an OSBPL2-deleted HeLa cell line using CRISPR/Cas9 technology, screened differentially expressed genes and examined the transcriptional regulation of SQLE using a dual-luciferase reporter gene. RNA-seq analysis showed that SQLE was upregulated significantly and the dual luciferase reporter gene assay revealed that two new functional transcription factor binding sites of Sp1 transcription factor (SP1) and sterol regulatory element-binding transcription factor 2 (SREBF2) in the SQLE promoter participated in the SQLE transcription and expression. In addition, we also observed that OSBPL2 deletion inhibited the AMPK signalling pathway and that the inhibition of AMPK signalling promoted SP1 and SREBF2 entry into the nuclear to upregulate SQLE expression. Therefore, these data support that OSBPL2 deficiency upregulates SQLE expression and increases the accumulation of cholesterol and cholesteryl ester by suppressing AMPK signalling, which provides new evidence of the connection between OSBPL2 and cholesterol synthesis.
Collapse
Affiliation(s)
- Cui Zhang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Hongdu Zhang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Min Zhang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Changsong Lin
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Hongshun Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Jun Yao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Qinjun Wei
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China; The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yajie Lu
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Zhibin Chen
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guangqian Xing
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China; The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
70
|
Chen J, Li Q, Ma J. Maternal serum, placental, and umbilical venous blood irisin levels in intrahepatic cholestasis of pregnancy. J Matern Fetal Neonatal Med 2019; 34:2403-2410. [PMID: 31590596 DOI: 10.1080/14767058.2019.1667322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To explore the changes of serum, umbilical vein, placental irisin level, and the correlation between irisin level and relevant indicators in pregnant women with intrahepatic cholestasis of pregnancy (ICP), so as to provide a new perspective for in-depth studies on the causes and treatment of ICP. METHODS This cross-sectional case-control study method, the serum, umbilical venous blood irisin, liver, kidney function, lipid metabolism, and other indicators of 108 normal pregnant women, 64 patients with mild ICP, and 39 patients with severe ICP were compared, and the changes in the levels of oxidative stress and irisin were observed by dihydroethidium staining and immunohistochemistry. RESULTS The level of placental oxidative stress in severe ICP group and mild ICP group was significantly higher than that in normal pregnant women group, and the mild ICP group was significantly higher than that in severe ICP group (p < .05); the concentration of irisin in umbilical vein was significantly lower than that in peripheral blood; the serum irisin of normal pregnant women (918.51 ± 159.90 pg/ml) was significantly lower than that of pregnant women with mild ICP (1030.05 ± 137.98 pg/ml) and pregnant women with severe ICP (1094.34 ± 154.35 pg/ml). The concentration of irisin in umbilical vein blood of pregnant women with severe ICP (858.78 ± 97.42 pg/ml) was significantly higher than that of normal pregnant women (595.33 ± 162.70 pg/ml) and those with mild ICP (648.82 ± 164.81 pg/ml) (p < .05). Irisin was expressed in placental tissues of normal pregnant women group, mild ICP group and severe ICP group, and the differences were statistically significant in expression intensity of the three groups (χ2 = 19.959, p < .05). The irisin expression intensity in the ICP group was higher than that in the control group, and the irisin expression intensity in the ICP group was higher than that in the ICP group (β = 0.292; t = 3.063; p < .05). At the best cutoff level of 989.168 pg/ml, irisin accurately predicted ICP [AUC = 0.622 (95%CI 0.543-0.701, p < .05)] with sensitivity and specificity rates of 60.9 and 40.1%, respectively. CONCLUSION Irisin can reduce the level of oxidative stress and improve lipid metabolism in ICP patients during the pathophysiological process of ICP, and it is possible to become a new auxiliary factor of ICP diagnosis and indexing.
Collapse
Affiliation(s)
- Jiaqing Chen
- Sichuan Provincial Hospital for Women and Children, Chengdu, Sichuan, China
| | - Qin Li
- Sichuan China 81 Rehabilitation Center, Chengdu, Sichuan, China
| | - Junru Ma
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
71
|
Ye X, Shen Y, Ni C, Ye J, Xin Y, Zhang W, Ren Y. Irisin reverses insulin resistance in C2C12 cells via the p38-MAPK-PGC-1α pathway. Peptides 2019; 119:170120. [PMID: 31351089 DOI: 10.1016/j.peptides.2019.170120] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 12/27/2022]
Abstract
Insulin resistance (IR) is a fundamental pathogenic factor shared by a myriad of metabolic disorders, including obesity and type 2 diabetes. The mechanism of IR is usually accompanied by mitochondrial dysfunction. Irisin has been proposed to act as a hormone in the regulation of energy homeostasis and metabolism. However, the effects of irisin on IR and mitochondrial function have not yet been fully investigated. Here, our research shows that irisin increases glucose uptake in C2C12 myoblast cells via the p38-mitogen-activated protein kinase (MAPK)-PGC-1α pathway. Irisin can also enhance mitochondrial function and mitochondrial respiration. Moreover, irisin stimulates autophagy via PGC-1α. Collectively, these data provide basic evidence to support the therapeutic potential of irisin for IR, which may rely on p38-MAPK-PGC-1α pathway activation and enhance mitochondrial function.
Collapse
Affiliation(s)
- Xiao Ye
- Department of Endocrinology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310003, China
| | - YiMin Shen
- Department of Endocrinology, Second Affiliated Hospital, Medical School of Zhejiang University, Hangzhou 310003, China
| | - Chao Ni
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310003, China
| | - Jun Ye
- Department of Gastroenterology, Second Affiliated Hospital, Medical School of Zhejiang University, Hangzhou 310003, China
| | - Yubo Xin
- Department of Endocrinology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310003, China
| | - Wei Zhang
- Department of Endocrinology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310003, China; Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310003, China.
| | - YueZhong Ren
- Department of Endocrinology, Second Affiliated Hospital, Medical School of Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
72
|
Abstract
Studies have linked obesity, metabolic syndrome, type 2 diabetes, cardiovascular disease (CVD), nonalcoholic fatty liver disease (NAFLD) and dementia. Their relationship to the incidence and progression of these disease states suggests an interconnected pathogenesis involving chronic low-grade inflammation and oxidative stress. Metabolic syndrome represents comorbidities of central obesity, insulin resistance, dyslipidemia, hypertension and hyperglycemia associated with increased risk of type 2 diabetes, NAFLD, atherosclerotic CVD and neurodegenerative disease. As the socioeconomic burden for these diseases has grown signficantly with an increasing elderly population, new and alternative pharmacologic solutions for these cardiometabolic diseases are required. Adipose tissue, skeletal muscle and liver are central endocrine organs that regulate inflammation, energy and metabolic homeostasis, and the neuroendocrine axis through synthesis and secretion of adipokines, myokines, and hepatokines, respectively. These organokines affect each other and communicate through various endocrine, paracrine and autocrine pathways. The ultimate goal of this review is to provide a comprehensive understanding of organ crosstalk. This will include the roles of novel organokines in normal physiologic regulation and their pathophysiological effect in obesity, metabolic syndrome, type 2 diabetes, CVD, NAFLD and neurodegenerative disorders.
Collapse
Affiliation(s)
- Hye Soo Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Seoul, South Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
73
|
Zhang X, Hu C, Kong CY, Song P, Wu HM, Xu SC, Yuan YP, Deng W, Ma ZG, Tang QZ. FNDC5 alleviates oxidative stress and cardiomyocyte apoptosis in doxorubicin-induced cardiotoxicity via activating AKT. Cell Death Differ 2019; 27:540-555. [PMID: 31209361 PMCID: PMC7206111 DOI: 10.1038/s41418-019-0372-z] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/11/2019] [Accepted: 06/03/2019] [Indexed: 12/29/2022] Open
Abstract
Oxidative stress and cardiomyocyte apoptosis play critical roles in doxorubicin (DOX)-induced cardiotoxicity. Previous studies indicated that fibronectin type III domain-containing 5 (FNDC5) and its cleaved form, irisin, could preserve mitochondrial function and attenuate oxidative damage as well as cell apoptosis, however, its role in DOX-induced cardiotoxicity remains unknown. Our present study aimed to investigate the role and underlying mechanism of FNDC5 on oxidative stress and cardiomyocyte apoptosis in DOX-induced cardiotoxicity. Cardiomyocyte-specific FNDC5 overexpression was achieved using an adeno-associated virus system, and then the mice were exposed to a single intraperitoneal injection of DOX (15 mg/kg) to generate DOX-induced cardiotoxicity. Herein, we found that FNDC5 expression was downregulated in DOX-treated murine hearts and cardiomyocytes. Fndc5 deficiency resulted in increased oxidative damage and apoptosis in H9C2 cells under basal conditions, imitating the phenotype of DOX-induced cardiomyopathy in vitro, conversely, FNDC5 overexpression or irisin treatment alleviated DOX-induced oxidative stress and cardiomyocyte apoptosis in vivo and in vitro. Mechanistically, we identified that FNDC5/Irisin activated AKT/mTOR signaling and decreased DOX-induced cardiomyocyte apoptosis, and moreover, we provided direct evidence that the anti-oxidant effect of FNDC5/Irisin was mediated by the AKT/GSK3β/FYN/Nrf2 axis in an mTOR-independent manner. And we also demonstrated that heat shock protein 20 was responsible for the activation of AKT caused by FNDC5/Irisin. In line with the data in acute model, we also found that FNDC5/Irisin exerted beneficial effects in chronic model of DOX-induced cardiotoxicity (5 mg/kg, i.p., once a week for three times, the total cumulative dose is 15 mg/kg) in mice. Based on these findings, we supposed that FNDC5/Irisin was a potential therapeutic agent against DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Chun-Yan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Peng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Si-Chi Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Yu-Pei Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China.,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China. .,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China. .,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, PR China. .,Cardiovascular Research Institute of Wuhan University, 430060, Wuhan, PR China. .,Hubei Key Laboratory of Cardiology, 430060, Wuhan, PR China.
| |
Collapse
|
74
|
Li H, Shen J, Wu T, Kuang J, Liu Q, Cheng S, Pu S, Chen L, Li R, Li Y, Zou M, Zhang Z, Jiang W, Qu A, He J. Irisin Is Controlled by Farnesoid X Receptor and Regulates Cholesterol Homeostasis. Front Pharmacol 2019; 10:548. [PMID: 31191305 PMCID: PMC6546903 DOI: 10.3389/fphar.2019.00548] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/01/2019] [Indexed: 02/05/2023] Open
Abstract
Objective The aim of this study was to investigate whether the nuclear receptor farnesoid X receptor (FXR) could regulate FNDC5/Irisin expression and the role of Irisin in hyperlipidemia and atherosclerosis in ApoE-/- mice. Methods and Results We treated primary human hepatocytes, HepG2 cells, and Rhesus macaques with FXR agonist (CDCA, GW4064, and ivermectin). FNDC5 expression was highly induced by CDCA and GW4064 in hepatocytes, HepG2 cells, and the circulating level of Irisin increased in Rhesus macaques. Luciferase reporter and CHIP assays were used to determine whether FXR could regulate FNDC5 promoter activity. Irisin-ApoE-/- and ApoE-/- mice were used to study the metabolic function of Irisin in dyslipidemia and atherosclerosis. Irisin-ApoE-/- mice showed improved hyperlipidemia and alleviated atherosclerosis as compared with ApoE-/- mice. Irisin upregulated the expression of Abcg5/Abcg8 in liver and intestine, which increased the transport of biliary cholesterol and fecal cholesterol output. Conclusion Activation of FXR induces FNDC5 mRNA expression in human and increased the circulating level of Irisin in Rhesus macaques. FNDC5/Irisin is a direct transcriptional target of FXR. Irisin may be a novel therapeutic strategy for dyslipidemia and atherosclerosis.
Collapse
Affiliation(s)
- Hong Li
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Shen
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tong Wu
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiangying Kuang
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shihai Cheng
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyun Pu
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Chen
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Li
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanping Li
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Min Zou
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiyong Zhang
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Jiang
- State Key Laboratory of Biotherapy, Molecular Medicine Research Center - Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jinhan He
- Department of Pharmacy, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
75
|
Li X, Duan H, Liu Q, Umar M, Luo W, Yang X, Zhu J, Li M. Construction of a Pichia pastoris strain efficiently secreting irisin and assessment of its bioactivity in HepG2 cells. Int J Biol Macromol 2019; 124:60-70. [DOI: 10.1016/j.ijbiomac.2018.11.092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/11/2018] [Accepted: 11/11/2018] [Indexed: 12/17/2022]
|
76
|
Yang N, Zhang H, Gao X, Miao L, Yao Z, Xu Y, Wang G. Role of irisin in Chinese patients with hypothyroidism: an interventional study. J Int Med Res 2019; 47:1592-1601. [PMID: 30722716 PMCID: PMC6460594 DOI: 10.1177/0300060518824445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective Irisin is a myokine that greatly affects energy expenditure and systemic metabolism. While thyroid hormone is likely associated with irisin, a direct relationship remains to be fully elucidated. This study aimed to investigate plasma irisin levels in Chinese patients with hypothyroidism. Methods A total of 155 subjects were divided into the hypothyroidism group or the control group. Fifty-seven patients in the hypothyroidism group received levothyroxine treatment. Baseline irisin levels were measured in the two groups and post-treatment levels were measured in the hypothyroidism group. Results Irisin levels were significantly lower in the hypothyroidism group than in the control group. In the hypothyroidism group, irisin levels were positively associated with free triiodothyronine and free thyroxine levels, and negatively associated with thyrotropin levels. In the hypothyroidism group, irisin levels were significantly increased after levothyroxine treatment. Multiple linear regression models showed that total cholesterol and free thyroxine levels were the only significant predictors of serum irisin levels. Conclusions Irisin levels are decreased in patients with hypothyroidism. Our results suggest that decreased irisin levels are directly associated with reduced thyroid hormone levels. These values may be restored after levothyroxine treatment in Chinese patients with hypothyroidism.
Collapse
Affiliation(s)
- Ning Yang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Heng Zhang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xia Gao
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Li Miao
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhi Yao
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yuan Xu
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
77
|
Khorasani ZM, Bagheri RK, Yaghoubi MA, Chobkar S, Aghaee MA, Abbaszadegan MR, Sahebkar A. The association between serum irisin levels and cardiovascular disease in diabetic patients. Diabetes Metab Syndr 2019; 13:786-790. [PMID: 30641808 DOI: 10.1016/j.dsx.2018.11.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 11/29/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Cardiovascular disease is the most common cause of mortality and morbidity in diabetic patients. Insulin resistance has been shown to be reduced by the secretion of irisin from muscle and adipose tissues. This study was aimed at determining the relationship between serum irisin levels and angiographically defined coronary artery disease (CAD) in type II diabetic patients. METHODS In this case-control study, 30 diabetic subjects with angiographically defined CAD were compared with 30 age- and sex-matched diabetic subjects without CAD in terms of clinical and laboratory parameters including serum irisin levels. RESULTS Serum levels of Irisin were significantly higher in the diabetic group without CAD compared with the group with CAD (P = 0.048). Serum irisin levels showed a significant positive correlation with BMI (r = 0.374, P = 0.004) and fasting insulin (r = 0.303, P = 0.021), and a significant negative correlation with diabetes duration (r = -0.384, P = 0.002). Based on the results of the binary logistic regression model, circulating levels of irisin were associated with the presence of CAD in diabetes (p = 0.038) after adjusting for potential confounders. CONCLUSION Serum irisin levels were lower in the diabetic patients with cardiovascular complication compared with the uncomplicated diabetic patients. Therefore, additional larger scale studies are needed to determine the role of irisin in monitoring CAD in diabetic patients.
Collapse
Affiliation(s)
| | - Ramin Khameneh Bagheri
- Department of Cardiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ali Yaghoubi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Chobkar
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Monavvar Afzal Aghaee
- Department of Social Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
78
|
Hsieh IC, Ho MY, Wen MS, Chen CC, Hsieh MJ, Lin CP, Yeh JK, Tsai ML, Yang CH, Wu VCC, Hung KC, Wang CC, Wang CY. Serum irisin levels are associated with adverse cardiovascular outcomes in patients with acute myocardial infarction. Int J Cardiol 2018; 261:12-17. [PMID: 29657036 DOI: 10.1016/j.ijcard.2017.11.072] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/15/2017] [Accepted: 11/20/2017] [Indexed: 11/17/2022]
Abstract
Irisin, a recently identified myokine, regulates mitochondrial function and energy expenditure. The concentration of irisin is significantly altered after ST-elevation myocardial infarction (STEMI). We hypothesized that serum irisin concentration is associated with adverse cardiovascular outcomes after myocardial infarction. Serum irisin concentrations were measured using enzyme-linked immunosorbent assay (ELISA) in 399 patients 28d after the onset of STEMI in a prospective single-center cohort study. We assessed the association between irisin concentrations and adverse cardiovascular events during a 3-year follow-up. The excess risks of cardiovascular mortality, stroke, heart failure, and revascularization were predominantly seen among those with the highest concentrations of irisin, with concentrations higher than 75th percentile of the overall distribution had a ~4-fold increase in risk (hazard ratio=3.96, 95% confidence interval 1.55 to 10.11, P<0.01). Our findings showed that serum concentrations of irisin are elevated in post-STEMI patients with increased risk for adverse cardiovascular events. Novel therapies targeting irisin may represent a new direction in the treatment of STEMI.
Collapse
Affiliation(s)
- I-Chang Hsieh
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Ming-Yun Ho
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Ming-Shien Wen
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Chun-Chi Chen
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Ming-Jer Hsieh
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Chia-Pin Lin
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Jih-Kai Yeh
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Ming-Lung Tsai
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Chia-Hung Yang
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Victor Chien-Chia Wu
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Kuo-Chun Hung
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Chun-Chieh Wang
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Chao-Yung Wang
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan.
| |
Collapse
|
79
|
Belviranlı M, Okudan N. Exercise training increases cardiac, hepatic and circulating levels of brain-derived neurotrophic factor and irisin in young and aged rats. Horm Mol Biol Clin Investig 2018; 36:hmbci-2018-0053. [PMID: 30367793 DOI: 10.1515/hmbci-2018-0053] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/11/2018] [Indexed: 01/01/2023]
Abstract
Background The objective of study was to examine the impacts of exercise training on cardiac, hepatic and plasma brain-derived neurotrophic factor (BDNF) and irisin levels in young and aged rats. Materials and methods Four-month-old (young) and 20-month-old (aged) female rats performed exercise training consisting of voluntary wheel running for 12 weeks. BDNF and irisin levels were analyzed in the heart, liver and plasma samples by using commercially available enzyme-linked immunosorbent assay (ELISA) kits. Results Cardiac, hepatic and plasma BDNF levels were lower in the aged sedentary rats, than in the young exercised and aged exercised rats (p < 0.05). Heart, liver and plasma irisin concentrations were lower in the aged sedentary group than in the young sedentary, young exercised and aged exercised groups (p < 0.05) and regular exercise increased irisin levels in all the analyzed tissues when compared to the sedentary counterparts (p < 0.05). Conclusions The current results show that regular exercise improves aging-induced decrease in the cardiac, hepatic and plasma BNDF and irisin levels.
Collapse
Affiliation(s)
- Muaz Belviranlı
- Selçuk University, School of Medicine, Department of Physiology, Konya 42030, Turkey, Phone: 90 332 224 47 31, Fax: 90 332 224 48 08
| | - Nilsel Okudan
- Selçuk University, School of Medicine, Department of Physiology, Konya, Turkey
| |
Collapse
|
80
|
Bi J, Zhang J, Ren Y, Du Z, Li Q, Wang Y, Wei S, Yang L, Zhang J, Liu C, Lv Y, Wu R. Irisin alleviates liver ischemia-reperfusion injury by inhibiting excessive mitochondrial fission, promoting mitochondrial biogenesis and decreasing oxidative stress. Redox Biol 2018; 20:296-306. [PMID: 30388684 PMCID: PMC6216086 DOI: 10.1016/j.redox.2018.10.019] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/02/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
Current management of liver ischemia-reperfusion (I/R) injury is mainly based on supportive care and no specific treatment is available. Irisin, a recently identified hormone, plays pivotal roles in energy expenditure and oxidative metabolism; however, it remains unknown whether irisin has any protective effects on hepatic I/R injury. In this study, we found that serum and liver irisin levels were markedly decreased at 24 h after hepatic I/R. Treatment with exogenous irisin improved liver function, reduced liver necrosis and cell apoptosis, and relieved inflammatory response after hepatic I/R. Meanwhile, exogenous irisin markedly inhibited mitochondrial fission related protein dynamin related protein 1 (drp-1) and fission 1 (Fis-1) expression in hepatic I/R. Additionally, treatment with exogenous irisin increased mitochondrial content and increased mitochondrial biogenesis related peroxisome proliferative activated receptor-γ (PPARγ) co-activator 1α (PGC-1α) and mitochondrial transcription factor (TFAM) expression. Furthermore, irisin decreased oxidative stress by upregulating uncoupling proteins (UCP) 2 expression in hepatic I/R. The results reveal that treatment with exogenous irisin alleviated hepatic I/R injury by restraining mitochondrial fission, promoting mitochondrial biogenesis and relieving oxidative stress. Irisin treatment appears to be a novel and promising therapeutic approach for hepatic I/R injury. Irisin protects hepatocytes against ischemia/reperfusion (I/R)-induced injury. Irisin inhibits excessive mitochondrial fission after hepatic I/R. Irisin promotes mitochondrial biogenesis after hepatic I/R. Irisin reduces oxidative stress after hepatic I/R.
Collapse
Affiliation(s)
- Jianbin Bi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Jia Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yifan Ren
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zhaoqing Du
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Qingshan Li
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yue Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Shasha Wei
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China
| | - Lifei Yang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chang Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, China; Institute of Advanced Surgical Technology and Engineering, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
81
|
Liu J, Song N, Huang Y, Chen Y. Irisin inhibits pancreatic cancer cell growth via the AMPK-mTOR pathway. Sci Rep 2018; 8:15247. [PMID: 30323244 PMCID: PMC6189061 DOI: 10.1038/s41598-018-33229-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 09/25/2018] [Indexed: 01/10/2023] Open
Abstract
Irisin, a recently identified myokine that is released from skeletal muscle following exercise, regulates body weight and influences various metabolic diseases such as obesity and diabetes. In this study, human recombinant nonglycosylated P-irisin (expressed in Escherichia coli prokaryote cell system) or glycosylated E-irisin (expressed in Pichia pastoris eukaryote cell system) were compared to examine the role of recombinant irisin against pancreatic cancer (PC) cells lines, MIA PaCa-2 and Panc03.27. MTT [3-(4, 5-dimethylthiazol-2-yl)-2, 5-di phenyltetrazolium bromide] and cell colony formation assays revealed that irisin significantly inhibited the growth of MIA PaCa-2 and Panc03.27 in a dose-dependent manner. Irisin also induced G1 arrest in both cell lines. Scratch wound healing and transwell assays revealed that irisin also inhibited the migration of PC cells. Irisin reversed the activity of epithelial–mesenchymal transition (EMT) while increasing E-cadherin expression and reducing vimentin expression. Irisin activated the adenosine monophosphate-activated protein kinase (AMPK) pathway and suppressed the mammalian target of rapamycin (mTOR) signaling. Besides, our results suggest that irisin receptors exist on the surface of human MIA PaCa-2 and Panc03.27 cells. Our results clearly demonstrate that irisin suppressed PC cell growth via the activation of AMPK, thereby downregulating the mTOR pathway and inhibiting EMT of PC cells.
Collapse
Affiliation(s)
- Jiayu Liu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qianjin Street, Changchun, 130012, China.,School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Nannan Song
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qianjin Street, Changchun, 130012, China.,School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yibing Huang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qianjin Street, Changchun, 130012, China.,School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yuxin Chen
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qianjin Street, Changchun, 130012, China. .,School of Life Sciences, Jilin University, Changchun, 130012, China.
| |
Collapse
|
82
|
Mahgoub MO, D'Souza C, Al Darmaki RSMH, Baniyas MMYH, Adeghate E. An update on the role of irisin in the regulation of endocrine and metabolic functions. Peptides 2018; 104:15-23. [PMID: 29608940 DOI: 10.1016/j.peptides.2018.03.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/29/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
Abstract
Irisin is a novel myokine and adipokine that has gained much attention recently due to its mechanisms of action. Irisin is secreted following proteolytic cleavage of its precursor fibronectin type III domain containing 5 (FNDC5). Following its release, irisin exerts its major action by increasing the expression of mitochondrial uncoupling protein 1 (UCP 1), which facilitates the conversion of white adipose tissue (WAT) into beige adipose tissue. Irisin is distributed in various body tissues and several actions have been attributed to its presence in those tissues. It has been suggested that it plays a role in metabolic diseases, ageing, inflammation and neurogenesis. However, the circulating levels of irisin are modulated by several factors such as diet, obesity, exercise, pharmacological agents and different pathological conditions. In this review, we have discussed the mechanisms by which irisin influences the functions of different body systems and how external factors in turn affect the circulating level of irisin. In conclusion, modification of circulating irisin level may help in the management of a variety of endocrine and metabolic disorders.
Collapse
Affiliation(s)
- Mohamed Omer Mahgoub
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - Crystal D'Souza
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - Reem S M H Al Darmaki
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - May M Y H Baniyas
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates.
| |
Collapse
|
83
|
Bakhshalizadeh S, Amidi F, Shirazi R, Shabani Nashtaei M. Vitamin D3 regulates steroidogenesis in granulosa cells through AMP-activated protein kinase (AMPK) activation in a mouse model of polycystic ovary syndrome. Cell Biochem Funct 2018; 36:183-193. [PMID: 29676471 DOI: 10.1002/cbf.3330] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/20/2018] [Accepted: 03/04/2018] [Indexed: 01/11/2023]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder in reproductive-aged women. Hormonal abnormality caused by steroidogenesis disturbances appears to be the main culprit of the clinical picture in PCOS. Vitamin D3 could regulate steroidogenesis in granulosa cells, but the mechanism of action of vitamin D3 on steroidogenesis remains unknown. AMP-activated protein kinase (AMPK) has a modulating role in steroid hormone production. We investigated the effect of vitamin D3 on steroidogenesis in cultured granulosa cells of dehydroepiandrosterone-induced PCOS mice and studied the involvement of AMPK signalling pathway in the current process. Immunoblotting assay showed that vitamin D3 could increase phosphorylation of AMPK alpha and acetyl-CoA carboxylase, main substrate of AMPK. Vitamin D3 and 5-aminoimidazole-4-carboxamide-1-β-D-riboside or Aicar (AMPK activator) not only reduced gene expression of steroidogenic enzymes (P450scc or Cyp11a1, StAR, Cyp19a1 and 3B-HSD), but also reduced production of progesterone and 17B-estradiol assessed by radioimmunoassay. Pretreatment with compound C (AMPK inhibitor) decreased APMK phosphorylation and eliminated the effects of vitamin D3 and Aicar on steroidogenic enzymes expression and estradiol and progesterone production. This study showed that vitamin D3 has the main role in regulating of steroidogenesis in granulosa cells of mouse polycystic ovary through activation of the AMPK signalling pathway. SIGNIFICANCE OF THE STUDY Polycystic ovarian syndrome (PCOS) is an endocrine disorder of women in reproductive age. This disorder is partly related to disruption in steroidogenesis pathway and dysregulation of estradiol and progesterone production in granulosa cells of polycystic ovaries. Previously, we have shown that vitamin D3 could modulate steroidogenesis pathway in PCOS granulosa cells. In this study, we investigate the molecular mechanism of vitamin D3 in regulation of steroidogenesis pathway. We have shown that vitamin D3 has a modulating role in steroidogenesis pathway of granulosa cells by regulation of AMP-activated protein kinase (AMPK) as an underlying molecular mechanism in mouse polycystic ovary.
Collapse
Affiliation(s)
- Shabnam Bakhshalizadeh
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fardin Amidi
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shirazi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
84
|
Fatima SS, Khalid E, Ladak AA, Ali SA. Colostrum and mature breast milk analysis of serum irisin and sterol regulatory element-binding proteins-1c in gestational diabetes mellitus. J Matern Fetal Neonatal Med 2018; 32:2993-2999. [PMID: 29609490 DOI: 10.1080/14767058.2018.1454422] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Background: We aimed to evaluate irisin and SREBP-1c levels in serum, colostrum and mature breast milk in women with and without gestational diabetes (GDM); and to relate them with maternal glucose, lipid profile and weight status of babies. Methods: GDM positive women (n = 33) and normal glucose tolerant women (NGT) (n = 33) were recruited. Maternal blood samples were collected at 28th week of gestation and later at 6-week post-partum while breast milk samples of the lactating mothers were collected within 72 hours of birth (colostrum) and at 6 weeks post-partum (mature milk). Irisin and SREBP-1c levels were analyzed by commercially available ELISA kits for all maternal samples. Results: Lower levels of irisin were seen in serum, colostrum and mature breast milk of GDM females (p < .01). SREBP-1c profile showed a similar trend of low serum levels in GDM, however, they were undetectable in colostrum and mature breast milk. Weak to moderate correlations of serum irisin with BMI (r = 0.439; p < .001), GTT 0 hours (r = 0.403; p = .01), HbA1c (r = -0.312; p = .011), Fasting blood glucose (r = 0.992; p = .008), and baby weight at birth (r = 0.486; p < .001). Colostrum and mature breast milk irisin showed positive associations with baby weight at 6 weeks (r = 0.325; p = .017; r = 0.296; p = .022, respectively). Serum SREBP-1c at 6 weeks correlated with random blood glucose (r = 0.318; p = .009), and HbA1c (r= -0.292; p = .011). All correlations were lost once we adjusted for maternal BMI. Conclusions: Low irisin and SREBP1-c levels may favor development of GDM in pregnant subjects. Further, low mature breast milk levels may act as a continued stressor from fetal to infant life as long as breast-feeding is continued. Further studies are required to identify the mechanistic relationship between these biomarkers and GDM.
Collapse
Affiliation(s)
- Syeda Sadia Fatima
- a Department of Biological and Biomedical Sciences , Aga Khan University , Karachi , Pakistan
| | - Erum Khalid
- b Department of Obstetrics and Gynecology , Hamdard University , Karachi , Pakistan
| | | | - Syed Adnan Ali
- d Department of Statistics , University of Karachi , Karachi , Pakistan
| |
Collapse
|
85
|
Polyzos SA, Anastasilakis AD, Efstathiadou ZA, Makras P, Perakakis N, Kountouras J, Mantzoros CS. Irisin in metabolic diseases. Endocrine 2018; 59:260-274. [PMID: 29170905 DOI: 10.1007/s12020-017-1476-1] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Irisin is a myokine/adipokine induced by the exercise in mice and humans, which is proposed to induce "browning" of white adipose tissue, its primary target, thus increasing thermogenesis and energy expenditure. Since its identification, irisin has been linked to favorable effects on metabolic diseases, including obesity, type 2 diabetes mellitus (T2DM), lipid metabolism and cardiovascular disease (CVD), nonalcoholic fatty liver disease (NAFLD), polycystic ovary syndrome (PCOS), and metabolic bone diseases. Generally, despite the promising profile of irisin in rodents, its effects on human are less recognized. REVIEW Most, but not all studies show a positive association between irisin and indices of adiposity. In T2DM, NAFLD, and CVD, most observational studies reported lower irisin levels in patients than controls. Regarding metabolic bone diseases, irisin is positively associated with bone mineral density and strength in athletes, and inversely associated with osteoporotic fractures in postmenopausal osteoporosis. In PCOS, data remain largely conflicting. Irisin does not seem to be further reduced when two metabolic diseases, e.g., T2DM and NAFLD, or obesity and NAFLD exist though more data are needed. Furthermore, it seems that diverse confounders may have affected the results of different clinical studies. CONCLUSION Irisin remains an appealing molecule from a pathophysiological point of view and an appealing therapeutic target for metabolic diseases, albeit much research is still needed.
Collapse
Affiliation(s)
- Stergios A Polyzos
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | - Zoe A Efstathiadou
- Department of Endocrinology, Ippokration General Hospital, Thessaloniki, Greece
| | - Polyzois Makras
- Department of Endocrinology and Diabetes, 251 Hellenic Air Force General Hospital, Athens, Greece
| | - Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jannis Kountouras
- Second Medical Clinic, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
86
|
Satomi-Kobayashi S, Ishida T. Modulation of High-Density Lipoprotein Function via Cardiac Rehabilitation. J Atheroscler Thromb 2018; 25:128-130. [PMID: 29142155 PMCID: PMC5827082 DOI: 10.5551/jat.ed089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
| | - Tatsuro Ishida
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine
| |
Collapse
|
87
|
The role of exercise-induced myokines in regulating metabolism. Arch Pharm Res 2017; 41:14-29. [DOI: 10.1007/s12272-017-0994-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 11/21/2017] [Indexed: 12/25/2022]
|
88
|
Gizaw M, Anandakumar P, Debela T. A Review on the Role of Irisin in Insulin Resistance and Type 2 Diabetes Mellitus. J Pharmacopuncture 2017; 20:235-242. [PMID: 30151293 PMCID: PMC6104716 DOI: 10.3831/kpi.2017.20.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 10/23/2017] [Accepted: 11/03/2017] [Indexed: 01/14/2023] Open
Abstract
Irisin is a novel hormone like polypeptide that is cleaved and secreted by an unknown protease from fibronectin type III domain-containing protein 5 (FNDC5), a membrane- spanning protein and which is highly expressed in skeletal muscle, heart, adipose tissue, and liver. Since its discovery in 2012, it has been the subject of many researches due to its potent physiological role. It is believed that understanding irisin’s function may be the key to comprehend many diseases and their development. Irisin is a myokine that leads to increased energy expenditure by stimulating the ‘browning’ of white adipose tissue. In the first description of this hormone, increased levels of circulating irisin, which is cleaved from its precursor fibronectin type III domain-containing protein 5, were associated with improved glucose homeostasis by reducing insulin resistance. Irisin is a powerful messenger, sending the signal to determine the function of specific cells, like skeletal muscle, liver, pancreas, heart, fat and the brain. The action of irisin on different targeted tissues or organs in human being has revealed its physiological functions for promoting health or executing the regulation of variety of metabolic diseases. Numerous studies focus on the association of irisin with metabolic diseases which has gained great interest as a potential new target to combat type 2 diabetes mellitus and insulin resistance. Irisin is found to improve insulin resistance and type 2 diabetes by increasing sensitization of the insulin receptor in skeletal muscle and heart by improving hepatic glucose and lipid metabolism, promoting pancreatic β cell functions, and transforming white adipose tissue to brown adipose tissue. This review is a thoughtful attempt to summarize the current knowledge of irisin and its effective role in mediating metabolic dysfunctions in insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Mamo Gizaw
- Biochemistry Unit, Department of Biomedical Sciences, College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Pandi Anandakumar
- Biochemistry Unit, Department of Biomedical Sciences, College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Tolessa Debela
- Physiology Unit, Department of Biomedical Sciences, College of Health Sciences, Arsi University, Asella, Ethiopia
| |
Collapse
|
89
|
Khidr EG, Ali SS, Elshafey MM, Fawzy OA. Association of irisin and FNDC5 rs16835198 G > T gene polymorphism with type 2 diabetes mellitus and diabetic nephropathy. An Egyptian pilot study. Gene 2017; 626:26-31. [DOI: 10.1016/j.gene.2017.05.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/25/2017] [Accepted: 05/03/2017] [Indexed: 12/29/2022]
|
90
|
Perakakis N, Triantafyllou GA, Fernández-Real JM, Huh JY, Park KH, Seufert J, Mantzoros CS. Physiology and role of irisin in glucose homeostasis. Nat Rev Endocrinol 2017; 13:324-337. [PMID: 28211512 PMCID: PMC5878942 DOI: 10.1038/nrendo.2016.221] [Citation(s) in RCA: 381] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Irisin is a myokine that leads to increased energy expenditure by stimulating the 'browning' of white adipose tissue. In the first description of this hormone, increased levels of circulating irisin, which is cleaved from its precursor fibronectin type III domain-containing protein 5, were associated with improved glucose homeostasis by reducing insulin resistance. Consequently, several studies attempted to characterize the role of irisin in glucose regulation, but contradictory results have been reported, and even the existence of this hormone has been questioned. In this Review, we present the current knowledge on the physiology of irisin and its role in glucose homeostasis. We describe the mechanisms involved in the synthesis, secretion, circulation and regulation of irisin, and the controversies regarding the measurement of irisin. We also discuss the direct effects of irisin on glucose regulatory mechanisms in different organs, the indirect effects and interactions with other hormones, and the important open questions with regard to irisin in those organs. Finally, we present the results from animal interventional studies and from human clinical studies investigating the association of irisin with obesity, insulin resistance, type 2 diabetes mellitus and the metabolic syndrome.
Collapse
Affiliation(s)
- Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Georgios A Triantafyllou
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - José Manuel Fernández-Real
- Department of Endocrinology, Hospital of Girona Doctor Josep Trueta, University of Girona, Emili Grahit Street, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avinguda de França, 17007 Girona, Spain
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, 77, Yongbong-ro, Buk-gu, Gwangju 61186, South Korea
| | - Kyung Hee Park
- Department of Family Medicine, Hallym University Sacred Heart Hospital, Hallym University, Gyeonggi-do 431070, South Korea
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Faculty of Medicine, University of Freiburg, 55 Hugstetter Street, 79106 Freiburg, Germany
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
- Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, 150 South Huntington Avenue, Boston, Massachusetts 02130, USA
| |
Collapse
|
91
|
Li DJ, Li YH, Yuan HB, Qu LF, Wang P. The novel exercise-induced hormone irisin protects against neuronal injury via activation of the Akt and ERK1/2 signaling pathways and contributes to the neuroprotection of physical exercise in cerebral ischemia. Metabolism 2017; 68:31-42. [PMID: 28183451 DOI: 10.1016/j.metabol.2016.12.003] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/18/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Irisin is a novel exercise-induced myokine involved in the regulation of adipose browning and thermogenesis. In this study, we investigated the potential role of irisin in cerebral ischemia and determined whether irisin is involved in the neuroprotective effect of physical exercise in mice. MATERIALS AND METHODS The middle cerebral artery occlusion (MCAO) model was used to produce cerebral ischemia in mice. First, the plasma irisin levels and changes in expression of the irisin precursor protein FNDC5 in skeletal muscle were determined post ischemic stroke. Second, the association between plasma irisin levels and the neurological deficit score, brain infarct volume, or plasma concentrations of tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1β in mice with MCAO were evaluated. Third, the therapeutic effect of irisin on ischemic brain injury was evaluated in vivo and in vitro. Recombinant irisin was injected directly into the tail vein 30min after the MCAO operation, and then the effects of irisin treatment on brain infarct volume, neurological deficit, neuroinflammation, microglia activation, monocyte infiltration, oxidative stress and intracellular signaling pathway activation (Akt and ERK1/2) were measured. Irisin was also administered in cultured PC12 neuronal cells with oxygen and glucose deprivation (OGD). Finally, to assess the potential involvement of irisin in the neuroprotection of physical exercise, mice were exercised for 2weeks and an irisin neutralizing antibody was injected into these mice to block irisin 1h before the MCAO operation. RESULTS The plasma irisin concentration and intramuscular FNDC5 protein expression decreased after ischemic stroke. Plasma irisin levels were negatively associated with brain infarct volume, the neurological deficit score, plasma TNF-α and plasma IL-6 concentrations. In OGD neuronal cells, irisin protected against cell injury. In mice with MCAO, irisin treatment reduced the brain infarct volume, neurological deficits, brain edema and the decline in body weight. Irisin treatment inhibited activation of Iba-1+ microglia, infiltration of MPO-1+ monocytes and expression of both TNF-α and IL-6 mRNA. Irisin significantly suppressed the levels of nitrotyrosine, superoxide anion and 4-hydroxynonenal (4-HNE) in peri-infarct brain tissues. Irisin treatment increased Akt and ERK1/2 phosphorylation, while blockade of Akt and ERK1/2 by specific inhibitors reduced the neuroprotective effects of irisin. Finally, the exercised mice injected with irisin neutralizing antibody displayed more severe neuronal injury than the exercised mice injected with control IgG. CONCLUSION Irisin reduces ischemia-induced neuronal injury via activation of the Akt and ERK1/2 signaling pathways and contributes to the neuroprotective effect of physical exercise against cerebral ischemia, suggesting that irisin may be a factor linking metabolism and cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| | - Yong-Hua Li
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hong-Bin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Le-Feng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Pei Wang
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
92
|
Shao L, Li H, Chen J, Song H, Zhang Y, Wu F, Wang W, Zhang W, Wang F, Li H, Tang D. Irisin suppresses the migration, proliferation, and invasion of lung cancer cells via inhibition of epithelial-to-mesenchymal transition. Biochem Biophys Res Commun 2016; 485:598-605. [PMID: 27986567 DOI: 10.1016/j.bbrc.2016.12.084] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 12/29/2022]
Abstract
Irisin is involved in promoting metabolism, immune regulation, and affects chronic inflammation in many systemic diseases, including gastric cancer. However, the role of irisin in lung cancer is not well characterized. To determine whether irisin has a protective effect against lung cancer, we cultured A549 and NCI-H446 lung cancer cells and treated them with irisin. We detected the proliferation by MTT assay, and assessed the migration and invasion of the cells by scratch wound healing assay and Tran-swell assay. The expression levels of epithelial-to-mesenchymal transition (EMT) markers and the related signaling pathways were detected by western blot analysis. Meanwhile, an inhibitor of PI3K was used to investigate the effect of irsin. Finally, the expression of Snail was detected. We demonstrated that irisin inhibits the proliferation, migration, and invasion of lung cancer cells, and has a novel role in mediating the PI3K/AKT pathway in the cells. Irisin can reverse the activity of EMT and inhibit the expression of Snail via mediating the PI3K/AKT pathway, which is a key regulator of Snail. These results revealed that irisin inhibited EMT and reduced the invasion of lung cancer cells via the PI3K/AKT/Snail pathway.
Collapse
Affiliation(s)
- Lei Shao
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China; Jinan Central Hospital Affiliated to Shandong University, Jinan, 250012, PR China
| | - Huanjie Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, 250012, PR China
| | - Jian Chen
- Jinan Central Hospital Affiliated to Shandong University, Jinan, 250012, PR China
| | - Haibo Song
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China
| | - Yuzhu Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China
| | - Fei Wu
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China
| | - Wenjuan Wang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China
| | - Wen Zhang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China
| | - Fang Wang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China
| | - Hui Li
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China
| | - Dongqi Tang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, PR China.
| |
Collapse
|