101
|
Ma J, Chen K. The role of Irisin in multiorgan protection. Mol Biol Rep 2021; 48:763-772. [PMID: 33389537 DOI: 10.1007/s11033-020-06067-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/03/2020] [Indexed: 02/08/2023]
Abstract
Physical exercise is an effective strategy for improving human health. Various organs, including the heart, lung and kidney, can benefit from exercise. However, the underlying molecular mechanisms by which exercise protects organs remain unknown. Irisin, a myokine secreted from muscle in response to exercise, has attracted increased attention from researchers. The role of irisin in multiorgan protection has been gradually revealed, and this muscle-derived circulating factor is regarded as an essential bridge linking exercise and organ health. The mechanisms by which irisin protects diverse organs are different. Here, we review the research progress on the multiorgan protective effects of irisin and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, People's Republic of China
| | - Ken Chen
- Department of Cardiology, Chongqing Renji Hospital, University of Chinese Academy of Sciences, Chongqing, 400062, People's Republic of China. .,Department of Cardiology, The Fifth People's Hospital of Chongqing, Chongqing, 400062, People's Republic of China.
| |
Collapse
|
102
|
Basini G, Bussolati S, Iannarelli M, Ragionieri L, Grolli S, Ramoni R, Dodi A, Gazza F, Grasselli F. The myokine irisin: localization and effects in swine late medium and large antral ovarian follicle. Domest Anim Endocrinol 2021; 74:106576. [PMID: 33120167 DOI: 10.1016/j.domaniend.2020.106576] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 01/10/2023]
Abstract
Irisin is mainly synthesized by skeletal muscle tissue, where it is believed to be responsible for the benefits of exercise on metabolism and cardiovascular system. In adipose tissue, its best-known effect is the browning of white adipocytes, resulting in the increase of thermogenesis and energy expenditure. As it has been largely documented that metabolic dysfunctions can frequently be associated with reductions in fertility, the possible involvement of this molecule in the regulation of reproductive processes represents an issue to be addressed. On this basis, the first aim of this work was the evaluation of the presence of irisin in the swine ovary; then, we investigated the expression of the associated molecules FNDC5, PGC-1α, and PPAR-γ. To verify a potential modulatory role both on ovarian function and on redox status, cell growth, steroidogenesis, production of superoxide anion and nitric oxide, the nonenzymatic antioxidant scavengers, were assessed in vitro on granulosa cells treated with increasing concentrations of irisin (50, 100, and 150 ng/mL). The data collected demonstrate the presence of irisin in swine ovarian follicle. Moreover, the highest concentrations tested stimulated metabolic activity and inhibited cell proliferation (P < 0.05); the peptide exerted a biphasic effect on progesterone (P < 0.01) production and, at the highest concentrations, inhibited nitric oxide while stimulated the nonenzymatic antioxidant power (P < 0.05). Superoxide anion and estradiol 17β were unaffected. The demonstration of the local presence of irisin at the ovarian level and the highlighted effects allow us to qualify this molecule as a potential physiological regulator of follicular function.
Collapse
Affiliation(s)
- G Basini
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy.
| | - S Bussolati
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - M Iannarelli
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - L Ragionieri
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - S Grolli
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - R Ramoni
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - A Dodi
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - F Gazza
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - F Grasselli
- Dipartimento di Scienze Medico-Veterinarie, Università di Parma, Via del Taglio 10, 43126 Parma, Italy
| |
Collapse
|
103
|
Jiang S, Oh DS, Dorotea D, Son E, Kim DS, Ha H. Dojuksan ameliorates tubulointerstitial fibrosis through irisin-mediated muscle-kidney crosstalk. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 80:153393. [PMID: 33120292 DOI: 10.1016/j.phymed.2020.153393] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/06/2020] [Accepted: 10/16/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Sarcopenia progresses in chronic kidney disease (CKD) and is positively correlated with mortality in end-stage kidney disease patients. Circulating irisin, an exercise-induced myokine, gradually decreases during CKD stage progression. Irisin inhibits the progression of kidney fibrosis, which is the final common outcome of CKD. Our preliminary study with C2C12 cells showed that Dojuksan, a herbal decoction, increases the expression of PGC1α (a regulator of irisin) and FNDC5 (a precursor of irisin). HYPOTHESIS Dojuksan may increase circulating irisin and prevent the progression of kidney fibrosis. STUDY DESIGN AND METHODS Unilateral ureteral obstruction (UUO) was performed on seven-week-old male C57BL/6 mice to induce kidney tubulointerstitial fibrosis. Dojuksan (50, 100, or 200 mg/kg/day) or losartan (1.5 mg/kg/day), a standard clinical treatment for CKD, was administered orally one day prior to surgery and continued for seven days thereafter. To determine the role of irisin released from muscles, TGFβ-stimulated murine proximal tubular epithelial cells (mProx24 cells) were treated with conditioned media (CM) from Dojuksan-treated C2C12 muscle cells transfected with FNDC5 siRNA. RESULTS UUO mice exhibited muscle wasting along with progressive kidney injury. Similar to losartan, Dojuksan ameliorated kidney inflammation and fibrosis in UUO mice. Dojuksan, but not losartan, increased plasma irisin concentration in UUO mice. Dojuksan significantly increased basal FNDC5 expression and inhibited TNFα-induced and indoxyl sulfate-induced FNDC5 down-regulation in C2C12 cells. The TGFβ-induced collagen I (COL1) up-regulation in mProx24 cells was effectively inhibited by CM from C2C12 cells after Dojuksan treatment. Moreover, irisin inhibited TGFβ-induced COL1 in mProx24 cells, which was not affected by CM from C2C12 cells transfected with FNDC5 siRNA. CONCLUSION Dojuksan ameliorates kidney fibrosis through irisin-mediated muscle-kidney crosstalk, suggesting that Dojuksan may be used as an alternative therapeutic agent against CKD.
Collapse
Affiliation(s)
- Songling Jiang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Dal-Seok Oh
- The Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Eunjung Son
- The Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Dong-Seon Kim
- The Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
104
|
Juszczak F, Vlassembrouck M, Botton O, Zwakhals T, Decarnoncle M, Tassin A, Caron N, Declèves AE. Delayed Exercise Training Improves Obesity-Induced Chronic Kidney Disease by Activating AMPK Pathway in High-Fat Diet-Fed Mice. Int J Mol Sci 2020; 22:ijms22010350. [PMID: 33396267 PMCID: PMC7795787 DOI: 10.3390/ijms22010350] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
Exercise training is now recognized as an interesting therapeutic strategy in managing obesity and its related disorders. However, there is still a lack of knowledge about its impact on obesity-induced chronic kidney disease (CKD). Here, we investigated the effects of a delayed protocol of endurance exercise training (EET) as well as the underlying mechanism in obese mice presenting CKD. Mice fed a high-fat diet (HFD) or a low-fat diet (LFD) for 12 weeks were subsequently submitted to an 8-weeks EET protocol. Delayed treatment with EET in obese mice prevented body weight gain associated with a reduced calorie intake. EET intervention counteracted obesity-related disorders including glucose intolerance, insulin resistance, dyslipidaemia and hepatic steatosis. Moreover, our data demonstrated for the first time the beneficial effects of EET on obesity-induced CKD as evidenced by an improvement of obesity-related glomerulopathy, tubulo-interstitial fibrosis, inflammation and oxidative stress. EET also prevented renal lipid depositions in the proximal tubule. These results were associated with an improvement of the AMPK pathway by EET in renal tissue. AMPK-mediated phosphorylation of ACC and ULK-1 were particularly enhanced leading to increased fatty acid oxidation and autophagy improvement with EET in obese mice.
Collapse
Affiliation(s)
- Florian Juszczak
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium; (M.V.); (T.Z.); (M.D.); (A.-E.D.)
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (O.B.); (N.C.)
- Correspondence: ; Tel.: +32-65373580
| | - Maud Vlassembrouck
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium; (M.V.); (T.Z.); (M.D.); (A.-E.D.)
| | - Olivia Botton
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (O.B.); (N.C.)
| | - Thomas Zwakhals
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium; (M.V.); (T.Z.); (M.D.); (A.-E.D.)
| | - Morgane Decarnoncle
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium; (M.V.); (T.Z.); (M.D.); (A.-E.D.)
| | - Alexandra Tassin
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium;
| | - Nathalie Caron
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium; (O.B.); (N.C.)
| | - Anne-Emilie Declèves
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium; (M.V.); (T.Z.); (M.D.); (A.-E.D.)
| |
Collapse
|
105
|
Thome T, Kumar RA, Burke SK, Khattri RB, Salyers ZR, Kelley RC, Coleman MD, Christou DD, Hepple RT, Scali ST, Ferreira LF, Ryan TE. Impaired muscle mitochondrial energetics is associated with uremic metabolite accumulation in chronic kidney disease. JCI Insight 2020; 6:139826. [PMID: 33290279 PMCID: PMC7821598 DOI: 10.1172/jci.insight.139826] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/25/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic kidney disease (CKD) causes progressive skeletal myopathy involving atrophy, weakness, and fatigue. Mitochondria have been thought to contribute to skeletal myopathy; however, the molecular mechanisms underlying muscle metabolism changes in CKD are unknown. We employed a comprehensive mitochondrial phenotyping platform to elucidate the mechanisms of skeletal muscle mitochondrial impairment in mice with adenine-induced CKD. CKD mice displayed significant reductions in mitochondrial oxidative phosphorylation (OXPHOS), which was strongly correlated with glomerular filtration rate, suggesting a link between kidney function and muscle mitochondrial health. Biochemical assays uncovered that OXPHOS dysfunction was driven by reduced activity of matrix dehydrogenases. Untargeted metabolomics analyses in skeletal muscle revealed a distinct metabolite profile in CKD muscle including accumulation of uremic toxins that strongly associated with the degree of mitochondrial impairment. Additional muscle phenotyping found CKD mice experienced muscle atrophy and increased muscle protein degradation, but only male CKD mice had lower maximal contractile force. CKD mice had morphological changes indicative of destabilization in the neuromuscular junction. This study provides the first comprehensive evaluation of mitochondrial health in murine CKD muscle to our knowledge and uncovers several unknown uremic metabolites that strongly associate with the degree of mitochondrial impairment.
Collapse
Affiliation(s)
- Trace Thome
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Ravi A Kumar
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Sarah K Burke
- Department of Physical Therapy, College of Public Health and Health Professions
| | - Ram B Khattri
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Zachary R Salyers
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Rachel C Kelley
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Madeline D Coleman
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Demetra D Christou
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| | - Russell T Hepple
- Department of Physical Therapy, College of Public Health and Health Professions
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA.,Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| |
Collapse
|
106
|
A brief overview about the physiology of fibronectin type III domain-containing 5. Cell Signal 2020; 76:109805. [PMID: 33031934 DOI: 10.1016/j.cellsig.2020.109805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/03/2020] [Accepted: 10/04/2020] [Indexed: 01/10/2023]
|
107
|
Shen F, Zhao Y, Ding W, Liu K, Ren X, Zhang Q, Yu J, Hu Y, Zuo H, Guo M, Jin L, Gong M, Wu W, Gu X, Xu L, Yang F, Lu J. Autonomous climbing: An effective exercise mode with beneficial outcomes of aerobic exercise and resistance training. Life Sci 2020; 265:118786. [PMID: 33221346 DOI: 10.1016/j.lfs.2020.118786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
AIMS To assess the effects of three specific exercise training modes, aerobic exercise (A), resistance training (R) and autonomous climbing (AC), aimed at proposing a cross-training method, on improving the physical, molecular and metabolic characteristics of mice without many side effects. MATERIALS AND METHODS Seven-week-old male mice were randomly divided into four groups: control (C), aerobic exercise (A), resistance training (R), and autonomous climbing (AC) groups. Physical changes in mice were tracked and analysed to explore the similarities and differences of these three exercise modes. Histochemistry, quantitative real-time PCR (RT-PCR), western blot (WB) and metabolomics analysis were performed to identify the underlying relationships among the three training modes. KEY FINDINGS Mice in the AC group showed better body weight control, glucose and energy homeostasis. Molecular markers of myogenesis, hypertrophy, antidegradation and mitochondrial function were highly expressed in the muscle of mice after autonomous climbing. The serum metabolomics landscape and enriched pathway comparison indicated that the aerobic oxidation pathway (pentose phosphate pathway, galactose metabolism and fatty acid degradation) and amino acid metabolism pathway (tyrosine, arginine and proline metabolism) were significantly enriched in group AC, suggesting an increased muscle mitochondrial function and protein balance ability of mice after autonomous climbing. SIGNIFICANCE We propose a new exercise mode, autonomous climbing, as a convenient but effective training method that combines the beneficial effects of aerobic exercise and resistance training.
Collapse
Affiliation(s)
- Fei Shen
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Yu Zhao
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China; Department of Physical Education, Northwestern Polytechnical University, Xi'an, Shaanxi 710049, PR China
| | - Wubin Ding
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Kailin Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China
| | - Xiangyu Ren
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China
| | - Qiang Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China
| | - Jian Yu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Yepeng Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, PR China; Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Hui Zuo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Ling Jin
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China
| | - Mingkai Gong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China
| | - Wenhao Wu
- School of Chemistry and Material Science, Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, PR China
| | - Xuejiang Gu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, PR China.
| | - Fenglei Yang
- School of Chemistry and Material Science, Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, PR China.
| | - Jian Lu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, PR China.
| |
Collapse
|
108
|
Bi Y, Chen Y, Xin D, Liu T, He L, Kang Y, Pan C, Shen W, Lan X, Liu M. Effect of indel variants within the sorting nexin 29 (SNX29) gene on growth traits of goats. Anim Biotechnol 2020; 33:914-919. [PMID: 33208046 DOI: 10.1080/10495398.2020.1846547] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The sorting nexin 29 gene (SNX29) is a well-known regulator of myocyte differentiation and proliferation. In this work, two indels (17-bp and 21-bp) were identified in the goat SNX29 gene, and their effects on the growth traits of 1,759 Shaanbei white cashmere (SBWC) goats were analyzed. Both indels had three genotypes [homozygote wild type (II), heterozygote (ID), and homozygote mutation (DD)] and displayed medium genetic diversity (0.25 < polymorphism information content (PIC) < 0.50) in the population. The 17-bp indel was significantly associated with chest width (p = 0.009), body weight (p = 0.021), and chest depth (p = 0.032), with the II genotype dominant. The 21-bp indel was significantly associated with chest width (p = 0.001), chest depth (p = 4.8E-5), heart girth (p = 0.007), and hip width (p = 0.002). Because the two indels were in the upstream (17-bp) and intron (21-bp) regions of the SNX29 gene, transcription factor binding sites were predicted. The IRF5 and MYC could bind with the 17-bp indel and 21-bp indel sequences, respectively. This study indicates that SNX29 is a promising candidate gene that can be used to improve meat production in goat breeding.
Collapse
Affiliation(s)
- Yi Bi
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.,College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yuhan Chen
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Dongyun Xin
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Tingting Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Libang He
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yuxin Kang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuanying Pan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Weijun Shen
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Mei Liu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
109
|
Wang XH, Price SR. Going micro in CKD-related cachexia. Nephrol Dial Transplant 2020; 35:1462-1464. [PMID: 32073623 PMCID: PMC7473799 DOI: 10.1093/ndt/gfaa025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/14/2020] [Indexed: 12/25/2022] Open
Affiliation(s)
- Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, GA, USA
| | - S Russ Price
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA.,Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| |
Collapse
|
110
|
Shen W, Fan K, Zhao Y, Zhang J, Xie M. Stevioside inhibits unilateral ureteral obstruction-induced kidney fibrosis and upregulates renal PPARγ expression in mice. J Food Biochem 2020; 44:e13520. [PMID: 33047331 DOI: 10.1111/jfbc.13520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/24/2020] [Accepted: 09/24/2020] [Indexed: 01/02/2023]
Abstract
This study aimed to examine the inhibitory effect of stevioside on unilateral ureteral obstruction (UUO)-induced kidney fibrosis. The UUO mice were daily given 50-100 mg/kg stevioside by gavage for 14 days after the operation. The results showed that stevioside decreased the levels of blood urea nitrogen and renal hydroxyproline, severity of kidney fibrosis, and expressions of renal collagen I/III and α-smooth muscle actin proteins. Importantly, stevioside increased the expressions of renal peroxisome proliferator-activated receptor γ (PPARγ) and Smad7 proteins and level of renal glutathione peroxidase, decreased the expressions of renal nuclear factor kappa B (NF-κB), signal transducer and activator of transcription 3 (STAT3), p-STAT3, transforming growth factor-β1 (TGF-β1), Smad2/3, and p-Smad2/3 proteins, suggesting that the antifibrotic mechanisms are related to the activation of PPARγ and subsequent downregulations of NF-κB-mediated STAT3 and TGF-β1 expressions and inhibition of Smad-mediated signaling pathway. These findings provide an applied perspective of stevioside for kidney fibrosis. PRACTICAL APPLICATIONS: Stevioside is widely used in food products as a sweetener, and it has many beneficial biological effects, including antidiabetes, antihypertension, and renal protective action. Here, we provide a novel potential application of stevioside in the prevention and treatment of kidney fibrosis.
Collapse
Affiliation(s)
- Wei Shen
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Ke Fan
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Junyan Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Meilin Xie
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
111
|
Inhibition of ROCK2 alleviates renal fibrosis and the metabolic disorders in the proximal tubular epithelial cells. Clin Sci (Lond) 2020; 134:1357-1376. [PMID: 32490513 DOI: 10.1042/cs20200030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
Non-specific inhibition of Rho-associated kinases (ROCKs) alleviated renal fibrosis in the unilateral ureteral obstruction (UUO) model, while genetic deletion of ROCK1 did not affect renal pathology in mice. Thus, whether ROCK2 plays a role in renal tubulointerstitial fibrosis needs to be clarified. In the present study, a selective inhibitor against ROCK2 or genetic approach was used to investigate the role of ROCK2 in renal tubulointerstitial fibrosis. In the fibrotic kidneys of chronic kidney diseases (CKDs) patients, we observed an enhanced expression of ROCK2 with a positive correlation with interstitial fibrosis. In mice, the ROCK2 protein level was time-dependently increased in the UUO model. By treating CKD animals with KD025 at the dosage of 50 mg/kg/day via intraperitoneal injection, the renal fibrosis shown by Masson's trichrome staining was significantly alleviated along with the reduced expression of fibrotic genes. In vitro, inhibiting ROCK2 by KD025 or ROCK2 knockdown/knockout significantly blunted the pro-fibrotic response in transforming growth factor-β1 (TGF-β1)-stimulated mouse renal proximal tubular epithelial cells (mPTCs). Moreover, impaired cellular metabolism was reported as a crucial pathogenic factor in CKD. By metabolomics analysis, we found that KD025 restored the metabolic disturbance, including the impaired glutathione metabolism in TGF-β1-stimulated tubular epithelial cells. Consistently, KD025 increased antioxidative stress enzymes and nuclear erythroid 2-related factor 2 (Nrf2) in fibrotic models. In addition, KD025 decreased the infiltration of macrophages and inflammatory response in fibrotic kidneys and blunted the activation of macrophages in vitro. In conclusion, inhibition of ROCK2 may serve as a potential novel therapy for renal tubulointerstitial fibrosis in CKD.
Collapse
|
112
|
Chambers JM, Wingert RA. PGC-1α in Disease: Recent Renal Insights into a Versatile Metabolic Regulator. Cells 2020; 9:E2234. [PMID: 33022986 PMCID: PMC7601329 DOI: 10.3390/cells9102234] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) is perhaps best known as a master regulator of mitochondrial biogenesis and function. However, by virtue of its interactions as a coactivator for numerous nuclear receptors and transcription factors, PGC-1α also regulates many tissue-specific tasks that include adipogenesis, angiogenesis, gluconeogenesis, heme biosynthesis, thermogenesis, and cellular protection against degeneration. Knowledge about these functions continue to be discovered with ongoing research. Unsurprisingly, alterations in PGC-1α expression lead to a range of deleterious outcomes. In this review, we provide a brief background on the PGC-1 family with an overview of PGC-1α's roles as an adaptive link to meet cellular needs and its pathological consequences in several organ contexts. Among the latter, kidney health is especially reliant on PGC-1α. Thus, we discuss here at length how changes in PGC-1α function impact the states of renal cancer, acute kidney injury (AKI) and chronic kidney disease (CKD), as well as emerging data that illuminate pivotal roles for PGC-1α during renal development. We survey a new intriguing association of PGC-1α function with ciliogenesis and polycystic kidney disease (PKD), where recent animal studies revealed that embryonic renal cyst formation can occur in the context of PGC-1α deficiency. Finally, we explore future prospects for PGC-1α research and therapeutic implications for this multifaceted coactivator.
Collapse
Affiliation(s)
- Joseph M. Chambers
- College of Pharmacy, Natural and Health Sciences, Manchester University, Fort Wayne, IN 46845, USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
113
|
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: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
114
|
Yu H, Chen D, Oyebamiji O, Zhao YY, Guo Y. Expression correlation attenuates within and between key signaling pathways in chronic kidney disease. BMC Med Genomics 2020; 13:134. [PMID: 32957963 PMCID: PMC7504859 DOI: 10.1186/s12920-020-00772-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Compared to the conventional differential expression approach, differential coexpression analysis represents a different yet complementary perspective into diseased transcriptomes. In particular, global loss of transcriptome correlation was previously observed in aging mice, and a most recent study found genetic and environmental perturbations on human subjects tended to cause universal attenuation of transcriptome coherence. While methodological progresses surrounding differential coexpression have helped with research on several human diseases, there has not been an investigation of coexpression disruptions in chronic kidney disease (CKD) yet. METHODS RNA-seq was performed on total RNAs of kidney tissue samples from 140 CKD patients. A combination of differential coexpression methods were employed to analyze the transcriptome transition in CKD from the early, mild phase to the late, severe kidney damage phase. RESULTS We discovered a global expression correlation attenuation in CKD progression, with pathway Regulation of nuclear SMAD2/3 signaling demonstrating the most remarkable intra-pathway correlation rewiring. Moreover, the pathway Signaling events mediated by focal adhesion kinase displayed significantly weakened crosstalk with seven pathways, including Regulation of nuclear SMAD2/3 signaling. Well-known relevant genes, such as ACTN4, were characterized with widespread correlation disassociation with partners from a wide array of signaling pathways. CONCLUSIONS Altogether, our analysis reported a global expression correlation attenuation within and between key signaling pathways in chronic kidney disease, and presented a list of vanishing hub genes and disrupted correlations within and between key signaling pathways, illuminating on the pathophysiological mechanisms of CKD progression.
Collapse
Affiliation(s)
- Hui Yu
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
| | - Danqian Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, School of Life Sciences, Northwest University, Xi’an, 710069 Shaanxi China
| | | | - Ying-Yong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, School of Life Sciences, Northwest University, Xi’an, 710069 Shaanxi China
| | - Yan Guo
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
| |
Collapse
|
115
|
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: 84] [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
|
116
|
Wang FS, Kuo CW, Ko JY, Chen YS, Wang SY, Ke HJ, Kuo PC, Lee CH, Wu JC, Lu WB, Tai MH, Jahr H, Lian WS. Irisin Mitigates Oxidative Stress, Chondrocyte Dysfunction and Osteoarthritis Development through Regulating Mitochondrial Integrity and Autophagy. Antioxidants (Basel) 2020; 9:antiox9090810. [PMID: 32882839 PMCID: PMC7555738 DOI: 10.3390/antiox9090810] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022] Open
Abstract
Compromised autophagy and mitochondrial dysfunction downregulate chondrocytic activity, accelerating the development of osteoarthritis (OA). Irisin, a cleaved form of fibronectin type III domain containing 5 (FNDC5), regulates bone turnover and muscle homeostasis. Little is known about the effect of Irisin on chondrocytes and the development of osteoarthritis. This study revealed that human osteoarthritic articular chondrocytes express decreased level of FNDC5 and autophagosome marker LC3-II but upregulated levels of oxidative DNA damage marker 8-hydroxydeoxyguanosine (8-OHdG) and apoptosis. Intra-articular administration of Irisin further alleviated symptoms of medial meniscus destabilization, like cartilage erosion and synovitis, while improved the gait profiles of the injured legs. Irisin treatment upregulated autophagy, 8-OHdG and apoptosis in chondrocytes of the injured cartilage. In vitro, Irisin improved IL-1β-mediated growth inhibition, loss of specific cartilage markers and glycosaminoglycan production by chondrocytes. Irisin also reversed Sirt3 and UCP-1 pathways, thereby improving mitochondrial membrane potential, ATP production, and catalase to attenuated IL-1β-mediated reactive oxygen radical production, mitochondrial fusion, mitophagy, and autophagosome formation. Taken together, FNDC5 loss in chondrocytes is correlated with human knee OA. Irisin repressed inflammation-mediated oxidative stress and extracellular matrix underproduction through retaining mitochondrial biogenesis, dynamics and autophagic program. Our analyses shed new light on the chondroprotective actions of this myokine, and highlight the remedial effects of Irisin on OA development.
Collapse
Affiliation(s)
- Feng-Sheng Wang
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chung-Wen Kuo
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Yu-Shan Chen
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Shao-Yu Wang
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Huei-Jing Ke
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Pei-Chen Kuo
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chin-Huei Lee
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Jian-Ching Wu
- Biobank and Tissue Bank, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Wen-Bin Lu
- Institute of Biomedical Sciences, National Sun Yat-Sun University, Kaohsiung 804, Taiwan; (W.-B.L.); (M.-H.T.)
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-Sun University, Kaohsiung 804, Taiwan; (W.-B.L.); (M.-H.T.)
| | - Holger Jahr
- Department of Anatomy and Cell Biology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Department of Orthopedic Surgery, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
- Correspondence: (H.J.); (W.-S.L.); Tel.: +886-7-731-7123 (W.-S.L.)
| | - Wei-Shiung Lian
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (F.-S.W.); (C.-W.K.); (Y.-S.C.); (S.-Y.W.); (H.-J.K.); (P.-C.K.); (C.-H.L.)
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Correspondence: (H.J.); (W.-S.L.); Tel.: +886-7-731-7123 (W.-S.L.)
| |
Collapse
|
117
|
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: 2.0] [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
|
118
|
Si M, Wang Q, Li Y, Lin H, Luo D, Zhao W, Dou X, Liu J, Zhang H, Huang Y, Lou T, Hu Z, Peng H. Inhibition of hyperglycolysis in mesothelial cells prevents peritoneal fibrosis. Sci Transl Med 2020; 11:11/495/eaav5341. [PMID: 31167927 DOI: 10.1126/scitranslmed.aav5341] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
Progressive peritoneal fibrosis affects patients receiving peritoneal dialysis (PD) and has no reliable treatment. The mechanisms that initiate and sustain peritoneal fibrosis remain incompletely elucidated. To overcome these problems, we developed a strategy that prevents peritoneal fibrosis by suppressing PD-stimulated mesothelial-to-mesenchymal transition (MMT). We evaluated single-cell transcriptomes of mesothelial cells obtained from normal peritoneal biopsy and effluent from PD-treated patients. In cells undergoing MMT, we found cellular heterogeneity and intermediate transition states associated with up-regulation of enzymes involved in glycolysis. The expression of glycolytic enzymes was correlated with the development of MMT. Using gene expression profiling and metabolomics analyses, we confirmed that PD fluid induces metabolic reprogramming, characterized as hyperglycolysis, in mouse peritoneum. We found that transforming growth factor β1 (TGF-β1) can substitute for PD fluid to stimulate hyperglycolysis, suppressing mitochondrial respiration in mesothelial cells. Blockade of hyperglycolysis with 2-deoxyglucose (2-DG) inhibited TGF-β1-induced profibrotic cellular phenotype and peritoneal fibrosis in mice. We developed a triad of adeno-associated viruses that overexpressed microRNA-26a and microRNA-200a while inhibiting microRNA-21a to target hyperglycolysis and fibrotic signaling. Intraperitoneal injection of the viral triad inhibited the development of peritoneal fibrosis induced by PD fluid in mice. We conclude that hyperglycolysis is responsible for MMT and peritoneal fibrogenesis, and this aberrant metabolic state can be corrected by modulating microRNAs in the peritoneum. These results could provide a therapeutic strategy to combat peritoneal fibrosis.
Collapse
Affiliation(s)
- Meijun Si
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.,Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qianqian Wang
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.,Nephrology Division, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Yin Li
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Hongchun Lin
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Dan Luo
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Wenbo Zhao
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xianrui Dou
- Nephrology Division, Shunde Hospital of Southern Medical University, Foshan 528300, China
| | - Jun Liu
- Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hui Zhang
- Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yong Huang
- Division of Gastrointestinal Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Tanqi Lou
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hui Peng
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
119
|
Qi H, Cao Q, Liu Q. MicroRNA-183 exerts a protective role in lupus nephritis through blunting the activation of TGF-β/Smad/TLR3 pathway via reducing Tgfbr1. Exp Cell Res 2020; 394:112138. [PMID: 32535034 DOI: 10.1016/j.yexcr.2020.112138] [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] [Received: 05/10/2020] [Revised: 06/02/2020] [Accepted: 06/07/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE The role of microRNA (miR)-183 has been elucidated in systemic lupus erythematosus, while whether it is also engaged in the lupus nephritis (LN) development remains opaque. The intention of this study is to examine the relevance of miR-183 downregulation in the pathogenesis of LN. METHODS The expression of miR-183 was first detected in MRL/lpr mice at weeks 8 and 12, followed by the assessment the effects of miR-183 on renal fibrosis and inflammatory response after overexpression or silencing of miR-183 in mice with LN. We further overexpressed or knocked-down miR-183 in human renal glomerular endothelial cells (HRGECs), and detected the expression patterns of inflammatory factors and Vimentin and α-SMA in the cells. Differentially expressed genes in HRGECs overexpressing miR-183 by microarrays were intersected with targeting mRNAs of miR-183 predicted by bioinformatics websites. The effects of transforming growth factor beta receptor 1 (Tgfbr1) and TGF-β/Smad/TLR3 pathway on renal damage in mice were verified by rescue experiments. RESULTS miR-183 expression was notably lower in MRL/lpr mice, and increased miR-183 expression inhibited renal fibrosis and inflammatory response in mice with LN. Moreover, miR-183 inhibitor in HRGECs remarkably promoted the expression of Vimentin and α-SMA and the secretion of inflammatory factors. miR-183 protected the mouse kidney from pathological damages by targeting and inhibiting Tgfbr1 expression. CONCLUSION miR-183 inhibited the expression of Tgfbr1 by direct targeting to disrupt the TGF-β/Smad/TLR3 pathway, thus repressing renal fibrosis and the secretion of inflammatory factors in LN.
Collapse
Affiliation(s)
- Huimeng Qi
- Department of General Practice, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Qin Cao
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Qiang Liu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China.
| |
Collapse
|
120
|
Ren Y, Zhang J, Wang M, Bi J, Wang T, Qiu M, Lv Y, Wu Z, Wu R. Identification of irisin as a therapeutic agent that inhibits oxidative stress and fibrosis in a murine model of chronic pancreatitis. Biomed Pharmacother 2020; 126:110101. [PMID: 32199226 DOI: 10.1016/j.biopha.2020.110101] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Abnormal activation of pancreatic stellate cells (PSCs) plays a crucial role in the pathogenesis of chronic pancreatitis (CP). Irisin, an exercise-induced hormone, has been shown to mitigate liver fibrosis by inhibiting the activation of hepatic stellate cells. However, the effect of irisin in CP has not been evaluated. METHODS This study aimed to determine whether irisin is protective in CP. CP was induced by 6 IP injections of cerulein (50 μg/kg/body weight). HPSCs were treated with 5 ng/ml TGF-β1 as in vitro experiment. RESULTS Our results showed that repeated cerulein injection induced severe pancreatic injury and fibrosis in mice and the serum irisin level in cerulein-treated mice decreased as in CP patients. Excessive oxidative and ER stress was also present in the pancreas of cerulein-treated mice. Irisin treatment significantly alleviated pancreatic injury and fibrosis, which was associated with reduced oxidative and ER stress. In cultured PSCs, irisin directly inhibited TGF-β-induced α-SMA and collagen I expression. This effect appears to be mediated through downregulation of kindlin-2 and inhibition of the SMAD2/3 pathway. CONCLUSIONS Irisin alleviated pancreatic injury and fibrosis, which was associated with reduced oxidative and ER stress. Thus, irisin may offer therapeutic potential for patients with CP.
Collapse
Affiliation(s)
- Yifan Ren
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 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, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Mengzhou Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Jianbin Bi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Tao Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Minglong Qiu
- Department of Orthopedic, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zheng Wu
- 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, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
121
|
Jhee JH, Joo YS, Han SH, Yoo T, Kang S, Park JT. High muscle-to-fat ratio is associated with lower risk of chronic kidney disease development. J Cachexia Sarcopenia Muscle 2020; 11:726-734. [PMID: 32020762 PMCID: PMC7296269 DOI: 10.1002/jcsm.12549] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 12/03/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity, a known risk factor for chronic kidney disease (CKD), is generally assessed using body mass index (BMI). However, BMI may not effectively reflect body composition, and the impact of muscle-to-fat (MF) mass balance on kidney function has not been elucidated. This study evaluated the association between body muscle and fat mass balance, represented as the MF ratio, and incident CKD development. METHODS Data were retrieved from a prospective community-based cohort study (Korean Genome and Epidemiology Study). Muscle and fat mass were measured using multifrequency bioelectrical impedance analysis. The study endpoint was incident CKD (estimated glomerular filtration rate <60 mL/min/1.73 m2 in at least two or more consecutive measurements during the follow-up period). RESULTS Totally, 7682 participants were evaluated. Their mean age was 51.7 ± 8.7 years, and 48% of the subjects were men. During a median follow-up of 140.0 (70.0-143.0) months, 633 (8.2%) subjects developed incident CKD. When the association between body composition and incident CKD was investigated, multivariable Cox proportional hazard analysis revealed that increase in MF ratio was related with a decreased risk of CKD development [per 1 increase in MF ratio: hazard ratio (HR), 0.86; 95% confidence interval (CI), 0.77-0.96; P = 0.008]. This association was also maintained when MF ratio was dichotomized according to sex-specific median values (high vs. low: HR, 0.83; 95% CI, 0.70-0.98; P = 0.031). Analyses preformed in a propensity score matched group also revealed a similar decreased risk of incident CKD in high MF ratio participants (high vs. low: HR, 0.84; 95% CI, 0.71-0.98; P = 0.037). This relationship between MF ratio and incident CKD risk was consistently significant across subgroups stratified by age, sex, hypertension, estimated glomerular filtration rate categories, and proteinuria. Among different BMI groups (normal, overweight, and obese), the relationship between high MF ratio and lower incident CKD risk was significant only in overweight and obese subjects. CONCLUSIONS Lower fat mass relative to muscle mass may lower the risk of CKD development in individuals with normal renal function. This relationship seems more prominent in overweight and obese subjects than in normal weight subjects.
Collapse
Affiliation(s)
- Jong Hyun Jhee
- Division of Nephrology, Department of Internal MedicineGangnam Severance Hospital, Yonsei University College of MedicineSeoulKorea
- Department of Internal Medicine, Institute of Kidney Disease ResearchSeverance Hospital, Yonsei University College of MedicineSeoulKorea
| | - Young Su Joo
- Division of Nephrology, Department of Internal MedicineMyongji HospitalGoyangGyeonggi‐doKorea
- Department of Internal Medicine, Institute of Kidney Disease ResearchSeverance Hospital, Yonsei University College of MedicineSeoulKorea
| | - Seong Hyeok Han
- Department of Internal Medicine, Institute of Kidney Disease ResearchSeverance Hospital, Yonsei University College of MedicineSeoulKorea
| | - Tae‐Hyun Yoo
- Department of Internal Medicine, Institute of Kidney Disease ResearchSeverance Hospital, Yonsei University College of MedicineSeoulKorea
| | - Shin‐Wook Kang
- Department of Internal Medicine, Institute of Kidney Disease ResearchSeverance Hospital, Yonsei University College of MedicineSeoulKorea
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUSYonsei UniversitySeoulKorea
| | - Jung Tak Park
- Department of Internal Medicine, Institute of Kidney Disease ResearchSeverance Hospital, Yonsei University College of MedicineSeoulKorea
| |
Collapse
|
122
|
Liu Y, Fu Y, Liu Z, Shu S, Wang Y, Cai J, Tang C, Dong Z. Irisin is induced in renal ischemia-reperfusion to protect against tubular cell injury via suppressing p53. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165792. [PMID: 32251763 DOI: 10.1016/j.bbadis.2020.165792] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/14/2022]
Abstract
Renal ischemia-reperfusion is a major cause of acute kidney injury, a disease currently without effective treatments. Irisin was initially identified as an important factor produced by muscles to mediate the health benefits of exercise, and recent work has further suggested its protective effect against lung and liver injury. However, the role of Irisin in kidney diseases, including renal ischemia-reperfusion injury (IRI), remains unknown. In the present study, we found that the Irisin precursor, fibronectin type III domain-containing protein 5 (Fndc5), was induced in renal tubules in a mouse model of renal IRI and in cultured mouse renal proximal tubular cells subjected ATP depletion injury. Functionally, silencing Fndc5 in cultured proximal tubular cells increased the sensitivity to ATP depletion-induced apoptosis, whereas both Fndc5 overexpression and supplementation of recombinant Irisin alleviated ATP depletion-induced apoptosis. In vivo, administration of recombinant Irisin dramatically attenuated kidney dysfunction, tissue damage, tubular cell apoptosis, and inflammation during renal IRI in mice. Mechanistically, Irisin suppressed the activation of p53 in renal IRI, a critical factor in tubular cell death. Together, these results indicate that Irisin is induced in renal IRI as a protective mechanism for renal tubular cells, suggesting the therapeutic potential of recombinant Irisin in renal IRI and related kidney diseases.
Collapse
Affiliation(s)
- Yuxue Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zhiwen Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shaoqun Shu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ying Wang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
123
|
Wang B, Wang J, He W, Zhao Y, Zhang A, Liu Y, Hassounah F, Ma F, Klein JD, Wang XH, Wang H. Exogenous miR-29a Attenuates Muscle Atrophy and Kidney Fibrosis in Unilateral Ureteral Obstruction Mice. Hum Gene Ther 2020; 31:367-375. [PMID: 31950871 DOI: 10.1089/hum.2019.287] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Renal fibrosis leads to end-stage renal disease, but antifibrotic drugs are difficult to develop. Chronic kidney disease often results in muscle wasting, and thereby increases morbidity and mortality. In this work, adeno-associated virus (AAV)-mediated overexpressing miR-29a was hypothesized to counteract renal fibrosis and muscle wasting through muscle-kidney crosstalk in unilateral ureteral obstruction (UUO) mice. miR-29a level was downregulated in the kidney and skeletal muscle of UUO mice. The secretion of exosome-encapsulated miR-29a increased in cultured skeletal muscle satellite cells and HEK293 renal cells after stimulation with serum from UUO mice. This result was confirmed by qPCR and microRNA deep sequencing in the serum exosomes of mice with obstructed ureters. A recombinant AAV-miR-29a was generated to overexpress miR-29a and injected into the tibialis anterior muscle of the mice 2 weeks before UUO surgery. AAV-miR-29a abrogated the UUO-induced upregulation of YY1 and myostatin in skeletal muscles. Renal fibrosis was also partially improved in the UUO mice with intramuscular AAV-miR-29a transduction. AAV-miR-29a overexpression reversed the increase in transforming growth factor β, fibronectin, alpha-smooth muscle actin, and collagen 1A1 and 4A1 levels in the kidney of UUO mice. AAV-green fluorescent protein was applied to trace the AAV route in vivo, and fluorescence was significantly visible in the injected/uninjected muscles and in the kidneys. In conclusion, intramuscular AAV-miR-29a injection attenuates muscle wasting and ameliorates renal fibrosis by downregulating several fibrotic-related proteins in UUO mice.
Collapse
Affiliation(s)
- Bin Wang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, P.R. China.,Institute of Nephrology, Zhong Da Hospital, Southeast University, Nanjing, China.,Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Juan Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Department of Nephrology, Shanghai General Hosptial, Shanghai Jiaotong University, Shanghai, China
| | - Wei He
- Department of Gastroenterology, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Yajie Zhao
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Aiqing Zhang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Liu
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Division of Nephrology, The 3rd People's Hospital of Datong, Shanxi Medical University, Taiyuan, China
| | - Faten Hassounah
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Fuying Ma
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Janet D Klein
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Xiaonan H Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Haidong Wang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, P.R. China.,Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| |
Collapse
|
124
|
Anti-renal fibrosis and anti-inflammation effect of urolithin B, ellagitannin-gut microbial-derived metabolites in unilateral ureteral obstruction rats. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
125
|
Albrecht E, Schering L, Buck F, Vlach K, Schober HC, Drevon CA, Maak S. Irisin: Still chasing shadows. Mol Metab 2020; 34:124-135. [PMID: 32180552 PMCID: PMC7033458 DOI: 10.1016/j.molmet.2020.01.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Considerable uncertainty remains regarding the veracity of measuring myokine irisin more than seven years after its original description. Unresolved issues include the nature of transcription of the irisin precursor fibronectin type III domain containing 5 (FNDC5) gene across species, the reliability of irisin levels measured with commercial enzyme-linked immunosorbent assays (ELISAs), and the overall validity of the recently published reference values for human serum measured with quantitative mass spectrometry. We utilized multiple species and measures to evaluate the robustness of commonly used reagents and methods for reporting irisin. Methods Amplification of cDNA was used to assess the FNDC5 transcript patterns in humans and mice. The specificity and sensitivity of different irisin antibodies were examined via western blotting. Quantification of circulating native irisin was conducted with mass spectrometry using an absolute quantification peptide for irisin. Results We show that there is a greater transcript diversity of human FNDC5 than currently annotated, but no indication of the expression of transcripts leading to a truncated form of irisin. Available irisin antibodies still bind to patterns of unspecific serum proteins, which compromise reliable measurements of irisin with ELISAs. Absolute quantification of irisin with labeled peptides by mass spectrometry is an advanced method but requires a multi-step sample preparation introducing uncontrollable variations in the measurement. Conclusion Our data represent an explicit warning against measuring circulating irisin using available methods. Measuring irisin is akin to chasing shadows. Transcription pattern of the host FNDC5 gene is not conserved from mouse to human. Irisin antibodies detect neither circulating irisin nor FNDC5 in humans and mice. Sample preparation impairs exact quantification of irisin by mass spectrometry. Results on irisin levels in humans and mice are still unreliable.
Collapse
Affiliation(s)
- Elke Albrecht
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Lisa Schering
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Friedrich Buck
- Institute of Clinical Chemistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konrad Vlach
- Department of Internal Medicine I, Municipal Hospital Suedstadt Rostock, Rostock, Germany
| | - Hans-Christof Schober
- Department of Internal Medicine I, Municipal Hospital Suedstadt Rostock, Rostock, Germany
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Steffen Maak
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.
| |
Collapse
|
126
|
Kim MH, Leem YH. The effects of peripherally-subacute treatment with irisin on hippocampal dendritogenesis and astrocyte-secreted factors. Phys Act Nutr 2019; 23:32-35. [PMID: 32018344 PMCID: PMC7004566 DOI: 10.20463/jenb.2019.0029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 12/20/2019] [Indexed: 12/27/2022] Open
Abstract
[Purpose] Fibronectin type III domain containing 5 (FNDC5)/irisin is an exercise-induced myokine, which contributes to cognitive functions. However, the relationship between the neuroprotective effects of FNDC5/irisin and hippocampal dendritic remodeling and astrocyte-secreted factors remains unclear. Therefore, we explored whether subchronic recombinant irisin treatment affected hippocampal morphology and some astrocyte-derived molecules. [Methods] Mice were intraperitoneally injected with irisin (0.5 μg/kg/day) for seven days, followed by their sacrifice two days later. Hippocampal morphometric parameters were analyzed and pgc-1a, fndc5, bdnf, and some astrocyte-derived factors mRNA levels were measured. [Results] Dendritic length, arborization, and spine density were enhanced by irisin regimen in hippocampal CA1 and CA3 areas. Hippocampal pgc-1a, fndc5, and bdnf mRNA levels were significantly increased by irisin treatment. Moreover, hevin mRNA levels were significantly enhanced, whereas tgf-b1 levels downregulated by irisin treatment. [Conclusion] FNDC5/irisin has dendritogenic activity probably through hevin induction and TGF-β1 suppression.
Collapse
|
127
|
Ren YF, Wang MZ, Bi JB, Zhang J, Zhang L, Liu WM, Wei SS, Lv Y, Wu Z, Wu RQ. Irisin attenuates intestinal injury, oxidative and endoplasmic reticulum stress in mice with L-arginine-induced acute pancreatitis. World J Gastroenterol 2019; 25:6653-6667. [PMID: 31832004 PMCID: PMC6906211 DOI: 10.3748/wjg.v25.i45.6653] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/08/2019] [Accepted: 11/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is often associated with intestinal injury, which in turn exaggerates the progression of AP. Our recent study has shown that a low level of serum irisin, a novel exercise-induced hormone, is associated with poor outcomes in patients with AP and irisin administration protects against experimental AP. However, the role of irisin in intestinal injury in AP has not been evaluated. AIM To investigate the effect of irisin administration on intestinal injury in experimental AP. METHODS AP was induced in male adult mice by two hourly intraperitoneal injections of L-arginine. At 2 h after the last injection of L-arginine, irisin (50 or 250 μg/kg body weight) or 1 mL normal saline (vehicle) was administered through intraperitoneal injection. The animals were sacrificed at 72 h after the induction of AP. Intestinal injury, apoptosis, oxidative and endoplasmic reticulum (ER) stress were evaluated. RESULTS Administration of irisin significantly mitigated intestinal damage, reduced apoptosis, and attenuated oxidative and ER stress in AP mice. In addition, irisin treatment also effectively downregulated serum tumor necrosis factor-alpha and interleukin-6 levels and alleviated injury in the pancreas, liver and lung of AP mice. CONCLUSION Irisin-mediated multiple physiological events attenuate intestinal injury following an episode of AP. Irisin has a great potential to be further developed as an effective treatment for patients with AP.
Collapse
Affiliation(s)
- Yi-Fan Ren
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Meng-Zhou Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Jian-Bin Bi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Jia Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Lin Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Wu-Ming Liu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Sha-Sha Wei
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Rong-Qian Wu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
128
|
Wu J, Dong J, Verzola D, Hruska K, Garibotto G, Hu Z, Mitch WE, Thomas SS. Signal regulatory protein alpha initiates cachexia through muscle to adipose tissue crosstalk. J Cachexia Sarcopenia Muscle 2019; 10:1210-1227. [PMID: 31507080 PMCID: PMC6903446 DOI: 10.1002/jcsm.12459] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/24/2019] [Accepted: 05/14/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Muscle wasting from chronic kidney disease (CKD) or from defective insulin signalling results in morbidity and, ultimately, mortality. We have identified an endogenous mediator of insulin resistance, signal regulatory protein alpha (SIRPα), which leads to cachexia in mice and is associated with cachexia in patients with CKD. METHODS We assessed insulin signalling and mechanisms causing muscle atrophy plus white adipose tissue (WAT) metabolism in mouse models of CKD or acute diabetes (streptozotocin treatment). We then examined these factors in mice with global knockout (KO) of SIRPα and sought mediators of metabolic responses in muscle and adipose tissues of mice with either muscle-specific or adipose tissue-specific KO of SIRPα. Metabolic responses were confirmed in primary cultures of adipose cells. RESULTS In mice with CKD, SIRPα expression was increased in WAT (three-fold, P < 0.05), and this was associated with precursors of cachexia: 'pathologic browning', thermogenesis, and a two-fold activation of protein kinase A (P < 0.05 vs. control mice) plus loss of adipose tissue mass. In contrast, mice with SIRPα global KO and CKD or acute diabetes experienced improved insulin signalling and activation of pAkt plus 'physiologic browning' of WAT. These mice avoided losses of muscle and adipose tissues and experienced a 31% improvement in survival (P < 0.05) than did wild-type mice with CKD. In muscle-specific SIRPα KO mice with CKD, we uncovered that serum SIRPα levels (P < 0.05) were suppressed and were associated with improved insulin signalling both in skeletal muscles and in WAT. These changes were accompanied by physiologic WAT browning. However, in adipose-specific SIRPα KO mice with CKD, levels of serum SIRPα were increased over two-fold (P < 0.05), while muscle losses were minimally inhibited. Clinical implications of SIRPα signalling are suggested by our findings that include increased SIRPα expression in muscle and adipose tissues (P < 0.05 vs. healthy controls) plus higher SIRPα levels in the serum of patients with CKD (2.4-fold, P=0.000017 vs. healthy controls). CONCLUSIONS Our results show that SIRPα plays an important role as an anti-insulin mediator regulating pathways to cachexia. In muscle-specific SIRPα KO, changes in SIRPα serum levels seem to improve insulin signalling in muscle and WAT, suggesting crosstalk between muscle and adipose tissue. Therefore, targeting SIRPα may prevent cachexia in patients with CKD or acute diabetes.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes, White/cytology
- Adipocytes, White/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/metabolism
- Animals
- Cachexia/etiology
- Cachexia/genetics
- Cachexia/metabolism
- Cell Communication
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Disease Models, Animal
- Gene Knockdown Techniques
- Humans
- Insulin/metabolism
- Male
- Mice
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- Phosphorylation
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Jiao Wu
- Selzman Institute for Kidney Health, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Jiangling Dong
- Selzman Institute for Kidney Health, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Daniela Verzola
- Nephrology Division, Department of MedicineUniversità degli Studi di GenovaGenoaItaly
| | - Keith Hruska
- Nephrology Division, Department of PediatricsWashington University School of MedicineSt. LouisMOUSA
| | - Giacomo Garibotto
- Nephrology Division, Department of MedicineUniversità degli Studi di GenovaGenoaItaly
| | - Zhaoyong Hu
- Selzman Institute for Kidney Health, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - William E. Mitch
- Selzman Institute for Kidney Health, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Sandhya S. Thomas
- Selzman Institute for Kidney Health, Department of MedicineBaylor College of MedicineHoustonTXUSA
- Nephrology Division, Department of MedicineMichael E. Debakey Veterans Affairs Medical CenterHoustonTXUSA
| |
Collapse
|
129
|
Jones DL, Haak AJ, Caporarello N, Choi KM, Ye Z, Yan H, Varelas X, Ordog T, Ligresti G, Tschumperlin DJ. TGFβ-induced fibroblast activation requires persistent and targeted HDAC-mediated gene repression. J Cell Sci 2019; 132:jcs.233486. [PMID: 31527052 DOI: 10.1242/jcs.233486] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/06/2019] [Indexed: 12/11/2022] Open
Abstract
Tissue fibrosis is a chronic disease driven by persistent fibroblast activation that has recently been linked to epigenetic modifications. Here, we screened a small library of epigenetic small-molecule modulators to identify compounds capable of inhibiting or reversing TGFβ-mediated fibroblast activation. We identified pracinostat, an HDAC inhibitor, as a potent attenuator of lung fibroblast activation and confirmed its efficacy in patient-derived fibroblasts isolated from fibrotic lung tissue. Mechanistically, we found that HDAC-dependent transcriptional repression was an early and essential event in TGFβ-mediated fibroblast activation. Treatment of lung fibroblasts with pracinostat broadly attenuated TGFβ-mediated epigenetic repression and promoted fibroblast quiescence. We confirmed a specific role for HDAC-dependent histone deacetylation in the promoter region of the anti-fibrotic gene PPARGC1A (PGC1α) in response to TGFβ stimulation. Finally, we identified HDAC7 as a key factor whose siRNA-mediated knockdown attenuates fibroblast activation without altering global histone acetylation. Together, these results provide novel mechanistic insight into the essential role HDACs play in TGFβ-mediated fibroblast activation via targeted gene repression.
Collapse
Affiliation(s)
- Dakota L Jones
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Andrew J Haak
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Kyoung M Choi
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhenqing Ye
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Huihuang Yan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University, Boston, MA 02118, USA
| | - Tamas Ordog
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Giovanni Ligresti
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
130
|
Choi HI, Park JS, Kim DH, Kim CS, Bae EH, Ma SK, Kim SW. PGC-1α Suppresses the Activation of TGF-β/Smad Signaling via Targeting TGFβRI Downregulation by let-7b/c Upregulation. Int J Mol Sci 2019; 20:5084. [PMID: 31614978 PMCID: PMC6829475 DOI: 10.3390/ijms20205084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 01/04/2023] Open
Abstract
TGF-β/Smad signaling is a major pathway in progressive fibrotic processes, and further studies on the molecular mechanisms of TGF-β/Smad signaling are still needed for their therapeutic targeting. Recently, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) was shown to improve renal fibrosis, making it an attractive target for chronic kidney diseases (CKDs). Here, we show the mechanism by which PGC-1α regulates the TGF-β/Smad signaling pathway using HK-2 cell lines stably overexpressing empty vector (mock cells) or human PGC1α (PGC1α cells). Stable PGC-1α overexpression negatively regulated the expression of TGF-β-induced epithelial-mesenchymal transition (EMT) markers (fibronectin, E-cadherin, vimentin, and α-SMA) and EMT-related transcription factors (Snail and Slug) compared to mock cells, inhibiting fibrotic progression. Interestingly, among molecules upstream of Smad2/3 activation, the gene expression of only TGFβRI, but not TGFβRII, was downregulated in PGC-1α cells. In addition, the downregulation of TGFβRI by PGC-1α was associated with the upregulation of let-7b/c, miRNA for which the 3' untranslated region (UTR) of TGFβRI contains a binding site. In conclusion, PGC-1α suppresses TGF-β/Smad signaling activation via targeting TGFβRI downregulation by let-7b/c upregulation.
Collapse
Affiliation(s)
- Hoon-In Choi
- Department of Internal Medicine, Chonnam National University Medical School, 42 Jebongro, Gwangju 61469, Korea.
| | - Jung Sun Park
- Department of Internal Medicine, Chonnam National University Medical School, 42 Jebongro, Gwangju 61469, Korea.
| | - Dong-Hyun Kim
- Department of Internal Medicine, Chonnam National University Medical School, 42 Jebongro, Gwangju 61469, Korea.
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School, 42 Jebongro, Gwangju 61469, Korea.
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, 42 Jebongro, Gwangju 61469, Korea.
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, 42 Jebongro, Gwangju 61469, Korea.
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, 42 Jebongro, Gwangju 61469, Korea.
| |
Collapse
|
131
|
Robinson KA, Baker LA, Graham-Brown MPM, Watson EL. Skeletal muscle wasting in chronic kidney disease: the emerging role of microRNAs. Nephrol Dial Transplant 2019; 35:1469-1478. [DOI: 10.1093/ndt/gfz193] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/27/2019] [Indexed: 12/17/2022] Open
Abstract
Abstract
Skeletal muscle wasting is a common complication of chronic kidney disease (CKD), characterized by the loss of muscle mass, strength and function, which significantly increases the risk of morbidity and mortality in this population. Numerous complications associated with declining renal function and lifestyle activate catabolic pathways and impair muscle regeneration, resulting in substantial protein wasting. Evidence suggests that increasing skeletal muscle mass improves outcomes in CKD, making this a clinically important research focus. Despite extensive research, the pathogenesis of skeletal muscle wasting is not completely understood. It is widely recognized that microRNAs (miRNAs), a family of short non-coding RNAs, are pivotal in the regulation of skeletal muscle homoeostasis, with significant roles in regulating muscle growth, regeneration and metabolism. The abnormal expression of miRNAs in skeletal muscle during disease has been well described in cellular and animal models of muscle atrophy, and in recent years, the involvement of miRNAs in the regulation of muscle atrophy in CKD has been demonstrated. As this exciting field evolves, there is emerging evidence for the involvement of miRNAs in a beneficial crosstalk system between skeletal muscle and other organs that may potentially limit the progression of CKD. In this article, we describe the pathophysiological mechanisms of muscle wasting and explore the contribution of miRNAs to the development of muscle wasting in CKD. We also discuss advances in our understanding of miRNAs in muscle–organ crosstalk and summarize miRNA-based therapeutics currently in clinical trials.
Collapse
Affiliation(s)
- Kate A Robinson
- Department of Infection Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, UK
| | - Luke A Baker
- Department of Health Sciences, College of Life Sciences, George Davies Centre, University of Leicester, Leicester, UK
| | - Matthew P M Graham-Brown
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital Leicester, Leicester, UK
| | - Emma L Watson
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital Leicester, Leicester, UK
| |
Collapse
|
132
|
Constitutive PGC-1α overexpression in skeletal muscle does not protect from age-dependent decline in neurogenesis. Sci Rep 2019; 9:12320. [PMID: 31444397 PMCID: PMC6707251 DOI: 10.1038/s41598-019-48795-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/29/2019] [Indexed: 01/26/2023] Open
Abstract
Aerobic exercise prevents age-dependent decline in cognition and hippocampal neurogenesis. The transcription factor peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α) mediates many of the exercise-induced benefits in skeletal muscle, including the release of factors into the circulation with neurotrophic effects. We use a transgenic mouse model with muscle-specific overexpression of PGC-1α to study the contribution of chronic muscle activation on exercise-induced effects on hippocampal neurogenesis in aging. Young and old transgenic and wild type animals of both sexes displayed a robust age-related reduction in newborn BrdU+-cells, immature neurons (DCX+-cells) and new mature BrdU+/NeuN+-neurons in the dentate gyrus. No differences were detected between genotypes or sexes. Analysis of serum proteins showed a tendency towards increased levels of myokines and reduced levels of pro-inflammatory cytokines for transgenic animals, but only musclin was found to be significantly up-regulated in transgenic animals. We conclude that constitutive muscular overexpression of PGC-1α, despite potent systemic changes, is insufficient for mimicking exercise-induced effects on hippocampal neurogenesis in aging. Continued studies are required to investigate the complex molecular mechanisms by which circulating signals could mediate exercise-induced effects on the central nervous system in disease and aging, with the aim of discovering new therapeutic possibilities for patients.
Collapse
|
133
|
Organ crosstalk: the potent roles of inflammation and fibrotic changes in the course of organ interactions. Inflamm Res 2019; 68:825-839. [PMID: 31327029 DOI: 10.1007/s00011-019-01271-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Organ crosstalk can be defined as the complex and mutual biological communication between distant organs mediated by signaling factors. Normally, crosstalk helps to coordinate and maintain homeostasis, but sudden or chronic dysfunction in any organ causes dysregulation in another organ. Many signal molecules, including cytokines and growth factors, are involved in the metabolic dysregulation, and excessive or inappropriate release of these molecules leads to organ dysfunction or disease (e.g., obesity, type 2 diabetes). AIM AND METHOD The aim of this review is to reveal the impact of organ crosstalk on the pathogenesis of diseases associated with organ interactions and the role of inflammatory and fibrotic changes in the organ dysfunction. After searching in MEDLINE, PubMed and Google Scholar databases using 'organ crosstalk' as a keyword, studies related to organ crosstalk and organ interaction were compiled and examined. CONCLUSION The organ crosstalk and the functional integration of organ systems are exceedingly complex processes. Organ crosstalk contributes to metabolic homeostasis and affects the inflammatory response, related pathways and fibrotic changes. As in the case of interactions between adipose tissue and intestine, stimulation of inflammatory mechanisms plays an active role in the development of diseases including insulin resistance, obesity, type 2 diabetes and hepatic steatosis. The increased level of knowledge about the 'crosstalk' between any organ and distant organs will facilitate the early diagnosis of the disease as well as the management of the treatment practices in the short- and long-term organ dysfunction.
Collapse
|
134
|
Peng S, Song C, Li H, Cao X, Ma Y, Wang X, Huang Y, Lan X, Lei C, Chaogetu B, Chen H. Circular RNA SNX29 Sponges miR-744 to Regulate Proliferation and Differentiation of Myoblasts by Activating the Wnt5a/Ca 2+ Signaling Pathway. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 16:481-493. [PMID: 31051333 PMCID: PMC6495097 DOI: 10.1016/j.omtn.2019.03.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/16/2022]
Abstract
Myogenesis is a complex and precisely orchestrated process that is highly regulated by several non-coding RNAs and signal pathways. Circular RNAs (circRNAs) represent a novel subclass of endogenous non-coding RNAs that have been identified in multiple species and tissues and play a vital role in post-transcriptional regulation in eukaryotes, but the precise molecular mechanism of action remains largely unknown. Here, we screened a candidate circRNA derived from the SNX29 gene, termed circSNX29 from our previous circRNAs sequencing data of bovine skeletal muscle, and further characterized its regulation and function during muscle development. The overexpression of circSNX29 facilitated myoblasts differentiation and inhibited cell proliferation. Computational analysis using RNAhybrid showed the potential for circSNX29 to sponge to miR-744 with nine potential binding sites. We tested this via a luciferase screening assay and found that circSNX29 directly interacted with miR-744 and downregulation of miR-744 efficiently reversed the suppression of Wnt5a and CaMKIIδ. Importantly, through the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enrichment analysis, Fluo-4, AM, cell permeant-calcium ion fluorescent probing, and western blotting assays, we found that overexpression of Wnt5a and circSNX29 activated the non-canonical Wnt5a/Ca2+ pathway. Overall, the evidence generated by our study elucidates the regulatory mechanisms of circSNX29 to function as a sponge for miRNA-744 in bovine primary myoblasts.
Collapse
Affiliation(s)
- Shujun Peng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chengchuang Song
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Hui Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiukai Cao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yilei Ma
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaogang Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yongzhen Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Buren Chaogetu
- Animal Disease Control Center of Haixi Mongolian and Tibetan Autonomous Prefecture, Delingha 817000, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
135
|
Xianyuan L, Wei Z, Yaqian D, Dan Z, Xueli T, Zhanglu D, Guanyi L, Lan T, Menghua L. Anti-renal fibrosis effect of asperulosidic acid via TGF-β1/smad2/smad3 and NF-κB signaling pathways in a rat model of unilateral ureteral obstruction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 53:274-285. [PMID: 30668407 DOI: 10.1016/j.phymed.2018.09.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 07/06/2018] [Accepted: 09/03/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Renal fibrosis is the most common pathway leading to end-stage renal disease. It is characterized by excess extracellular matrix (ECM) accumulation and renal tissue damage, subsequently leading to kidney failure. Asperulosidic acid (ASPA), a bioactive iridoid glycoside, exerts anti-tumor, anti-oxidant, and anti-inflammatory activities, but its effects on renal fibrosis induced by unilateral ureteral obstruction (UUO) have not yet been investigated. PURPOSE This study aimed to investigate the protective effect of ASPA on renal fibrosis induced by UUO, and to explore its pharmacological mechanism. METHODS Thirty-six Sprague-Dawley (SD) rats were randomly divided into six groups: sham group, UUO model group, three ASPA treatment groups (10, 20, and 40 mg/kg), and captopril group (20 mg/kg). Rats were administered vehicle, ASPA or captopril intraperitoneally once a day for 14 consecutive days. Urea nitrogen (BUN), uric acid (UA) and inflammatory factors in serum samples were evaluated on the 7th, 10th, and 14th day after renal fibrosis induction. In addition, the 12 h urine was collected to test the content of urinary protein (upro) on the 14th day. The obstructive renal tissues were collected for pathological analysis (hematoxylin and eosion (H&E) staining and Masson's Trichrome staining) and immunohistochemical analysis on the 14th day after renal fibrosis induction. The mRNA expression of related factors and the protein levels of smad2, smad3, and smad4 were measured in UUO-induced rats by real time PCR and Western blot, respectively. RESULTS The levels of BUN, UA, and upro were elevated in UUO-induced rats, but ASPA treatment improved renal function by reducing the levels of BUN, UA, and upro. The protein levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-6, as well as the mRNA levels of TNF-α, IL-1β, IL-6, monocyte chemoattractant protein-1 (MCP-1) and interferon-γ (IFN-γ), were decreased after ASPA administration (10, 20 and 40 mg/kg) in a dose-dependent manner. The ASPA exerted an alleviation effect on the inflammatory response through inhibition of nuclear factor-kappa B (NF-κB) pathway. In addition, reductions in α-smooth muscle actin (α-SMA), collagen III, and fibronectin expression were observed after ASPA administration at doses of 20 and 40 mg/kg. Furthermore, the renal expression of transforming growth factor-β1 (TGF-β1), smad2, smad3, and smad4 was down-regulated by ASPA treatment at doses of 20 and 40 mg/kg. CONCLUSION ASPA possessed protective effects on renal interstitial fibrosis in UUO-induced rats. These effects may be through inhibition of the activation of NF-κB and TGF-β1/smad2/smad3 signaling pathways.
Collapse
Affiliation(s)
- Lu Xianyuan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zou Wei
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics & Gynecology Research, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410008, Hunan, China.
| | - Dong Yaqian
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zhou Dan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Tong Xueli
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Dong Zhanglu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Liang Guanyi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Tang Lan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Liu Menghua
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
136
|
Mafra D, Gidlund EK, Borges NA, Magliano DC, Lindholm B, Stenvinkel P, von Walden F. Bioactive food and exercise in chronic kidney disease: Targeting the mitochondria. Eur J Clin Invest 2018; 48:e13020. [PMID: 30144313 DOI: 10.1111/eci.13020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 08/11/2018] [Accepted: 08/22/2018] [Indexed: 12/16/2022]
Abstract
Chronic kidney disease (CKD), which affects 10%-15% of the population, associates with a range of complications-such as cardiovascular disease, frailty, infections, muscle and bone disorders and premature ageing-that could be related to alterations of mitochondrial number, distribution, structure and function. As mitochondrial biogenesis, bioenergetics and the dynamic mitochondrial networks directly or indirectly regulate numerous intra- and extracellular functions, the mitochondria have emerged as an important target for interventions aiming at preventing or improving the treatment of complications in CKD. In this review, we discuss the possible role of bioactive food compounds and exercise in the modulation of the disturbed mitochondrial function in a uraemic milieu.
Collapse
Affiliation(s)
- Denise Mafra
- Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil.,Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Eva-Karin Gidlund
- Division of Molecular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Natália Alvarenga Borges
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - D'Angelo Carlo Magliano
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Bengt Lindholm
- Division of Renal Medicine, Department of Clinical Science Intervention and Technology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science Intervention and Technology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ferdinand von Walden
- Division of Pediatric Neurology, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
137
|
Kume S, Nagasu H, Nangaku M, Nishiyama A, Nakamoto H, Kashihara N. Summary of the 2018 ISN Frontiers Meeting: Kidney Disease and Cardiovascular Disease. Kidney Int Rep 2018. [PMCID: PMC6035142 DOI: 10.1016/j.ekir.2018.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
International Society of Nephrology (ISN) Frontiers meetings build on the success of the ISN Nexus and Forefronts series by bringing together basic scientists, clinicians, and practitioners in a unique setting. This new event was organized to make more innovative science available to a global audience by removing regional barriers in accessing the latest knowledge. The first ISN Frontiers meeting was organized in partnership between the Japanese Society of Nephrology and the Japanese Society for Dialysis Therapy, which was held in Tokyo in February 2018. The meeting focused on the topic “Kidney Disease & Cardiovascular Disease,” which covered a broad range of scientific and clinical fields, including nephrology, cardiovascular diseases, dialysis, transplantation, chronic kidney disease (CKD)–mineral bone disease (MBD), diabetes, pediatric nephrology, nutrition, pharmacology, and nursing. A total of 1584 active physicians and scientists from 64 countries attended the meeting, and a number of leading physician scientists from different and related disciplines of clinical and basic research described and reviewed recent discoveries. This report summarizes the main highlights of the meeting lectures.
Collapse
Affiliation(s)
- Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Shiga, Japan
- Correspondence: Shinji Kume, Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan.
| | - Hajime Nagasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, Okayama, Japan
| | - Masaomi Nangaku
- Department of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Hidetomo Nakamoto
- Department of General Internal Medicine, Saitama Medical University, Saitama, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Okayama, Japan
| |
Collapse
|
138
|
Zhu SZ, Szeto V, Bao MH, Sun HS, Feng ZP. Pharmacological approaches promoting stem cell-based therapy following ischemic stroke insults. Acta Pharmacol Sin 2018; 39:695-712. [PMID: 29671416 DOI: 10.1038/aps.2018.23] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/13/2018] [Indexed: 02/06/2023]
Abstract
Stroke can lead to long-term neurological deficits. Adult neurogenesis, the continuous generation of newborn neurons in distinct regions of the brain throughout life, has been considered as one of the appoaches to restore the neurological function following ischemic stroke. However, ischemia-induced spontaneous neurogenesis is not suffcient, thus cell-based therapy, including infusing exogenous stem cells or stimulating endogenous stem cells to help repair of injured brain, has been studied in numerous animal experiments and some pilot clinical trials. While the effects of cell-based therapy on neurological function during recovery remains unproven in randomized controlled trials, pharmacological agents have been administrated to assist the cell-based therapy. In this review, we summarized the limitations of ischemia-induced neurogenesis and stem-cell transplantation, as well as the potential proneuroregenerative effects of drugs that may enhance efficacy of cell-based therapies. Specifically, we discussed drugs that enhance proliferation, migration, differentiation, survival and function connectivity of newborn neurons, which may restore neurobehavioral function and improve outcomes in stroke patients.
Collapse
|
139
|
Uysal N, Yuksel O, Kizildag S, Yuce Z, Gumus H, Karakilic A, Guvendi G, Koc B, Kandis S, Ates M. Regular aerobic exercise correlates with reduced anxiety and incresed levels of irisin in brain and white adipose tissue. Neurosci Lett 2018; 676:92-97. [PMID: 29655944 DOI: 10.1016/j.neulet.2018.04.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 01/02/2023]
Abstract
We have recently shown that regular voluntary aerobic exercised rats have low levels of anxiety. Irisin is an exercise-induced myokine that is produced by many tissues; and the role it plays in anxiolytic behavior is unknown. In this study we aimed to investigate the correlation between anxiety like behavior and irisin levels following regular voluntary aerobic exercise in male mice. We've have shown that anxiety levels decreased in exercised mice, while irisin levels increased in the brain, brown adipose tissue, white adipose tissue, kidney, and pancreas tissues. No significant difference of irisin levels in the liver, muscle and serum were detected in the exercise group, when compared to controls. In addition, there was a strong positive correlation between brain irisin levels and activity in middle area of open field test and in the open arms of elevated plus maze test; both which are indicators of low anxiety levels. Our results suggest that decrease in anxiolytic behavior due to regular voluntary exercise may be associated with locally produced brain irisin. White adipose tissue irisin levels also correlated very strongly with low anxiety. However, no serum irisin increase was detected, ruling out the possibility of increased peripheral irisin levels affecting the brain via the bloodstream. Further research is necessary to explain the mechanisms of which peripheral and central irisin effects anxiety and the brain region affected.
Collapse
Affiliation(s)
- Nazan Uysal
- Department of Physiology, Dokuz Eylul University, School of Medicine, Izmir, Turkey.
| | - Oguz Yuksel
- Department of Sports Medicine, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | - Servet Kizildag
- College of Vocational School of Health Services, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | - Zeynep Yuce
- Department of Medical Biology and Genetics, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | - Hikmet Gumus
- Department of Physiology, Dokuz Eylul University, School of Medicine, Izmir, Turkey; Dokuz Eylul University, School of Sport Sciences and Technology, Izmir, Turkey
| | - Aslı Karakilic
- Department of Physiology, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | - Guven Guvendi
- Department of Physiology, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | - Basar Koc
- Department of Physiology, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | - Sevim Kandis
- Department of Physiology, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | - Mehmet Ates
- College of Vocational School of Health Services, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| |
Collapse
|