1
|
Song X, Liu G, Bin Y, Bai R, Liang B, Yang H. C1q/Tumor Necrosis Factor-Related Protein-9 Enhances Macrophage Cholesterol Efflux and Improves Reverse Cholesterol Transport via AMPK Activation. Biochem Genet 2025; 63:1620-1634. [PMID: 38600398 PMCID: PMC11929689 DOI: 10.1007/s10528-024-10761-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024]
Abstract
Cholesterol efflux from foam cells in atherosclerotic plaques is crucial for reverse cholesterol transport (RCT), an important antiatherogenic event. ATP-binding cassette (ABC) transporters, ABCA1 and ABCG1, are key receptors in the cholesterol efflux pathway. C1q/tumor necrosis factor-related protein-9 (CTRP9) is a newly discovered adipokine and exhibits an atheroprotective activity. However, the role of CTRP9 in RCT still remains unknown. In this work, we investigated the effect of subcutaneous administration of CTRP9 protein on RCT and atherosclerotic lesion formation in ApoE-/- mice fed with a high-fat diet. CTRP9-dependent regulation of cholesterol efflux and ABC transporters in RAW 264.7 foam cells was determined. Our results showed that CTRP9 protein decreased atherosclerotic lesions, increased cholesterol efflux, and upregulated liver ABCA1 and ABCG1 expression in ApoE-/- mice. CTRP9 treatment dose-dependently increased mRNA and protein expression of ABCA1, ABCG1, and LXR-α in RAW 264.7 foam cells. Moreover, the expression and phosphorylation of AMPK was potentiated upon CTRP9 treatment. Notably, CTRP9-induced cholesterol efflux and upregulation of ABCA, ABCG1, and LXR-α were impaired when AMPK was knocked down. AMPK depletion restored cholesterol accumulation in CTRP9-treated RAW 264.7 cells. Taken together, subcutaneous injection is an effective novel delivery route for CTRP9 protein, and exogenous CTRP9 can facilitate cholesterol efflux and promote RCT in an animal model of atherosclerosis. The atheroprotective activity of CTRP9 is mediated through the activation of AMPK signaling.
Collapse
Affiliation(s)
- Xiaosu Song
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Gaizhen Liu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Yunfei Bin
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Rui Bai
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Huiyu Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China.
| |
Collapse
|
2
|
Chen F, Sarver DC, Saqib M, Velez LM, Aja S, Seldin MM, Wong GW. Loss of CTRP10 results in female obesity with preserved metabolic health. eLife 2025; 13:RP93373. [PMID: 40126547 PMCID: PMC11932693 DOI: 10.7554/elife.93373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
Obesity is a major risk factor for type 2 diabetes, dyslipidemia, cardiovascular disease, and hypertension. Intriguingly, there is a subset of metabolically healthy obese (MHO) individuals who are seemingly able to maintain a healthy metabolic profile free of metabolic syndrome. The molecular underpinnings of MHO, however, are not well understood. Here, we report that CTRP10/C1QL2-deficient mice represent a unique female model of MHO. CTRP10 modulates weight gain in a striking and sexually dimorphic manner. Female, but not male, mice lacking CTRP10 develop obesity with age on a low-fat diet while maintaining an otherwise healthy metabolic profile. When fed an obesogenic diet, female Ctrp10 knockout (KO) mice show rapid weight gain. Despite pronounced obesity, Ctrp10 KO female mice do not develop steatosis, dyslipidemia, glucose intolerance, insulin resistance, oxidative stress, or low-grade inflammation. Obesity is largely uncoupled from metabolic dysregulation in female KO mice. Multi-tissue transcriptomic analyses highlighted gene expression changes and pathways associated with insulin-sensitive obesity. Transcriptional correlation of the differentially expressed gene (DEG) orthologs in humans also shows sex differences in gene connectivity within and across metabolic tissues, underscoring the conserved sex-dependent function of CTRP10. Collectively, our findings suggest that CTRP10 negatively regulates body weight in females, and that loss of CTRP10 results in benign obesity with largely preserved insulin sensitivity and metabolic health. This female MHO mouse model is valuable for understanding sex-biased mechanisms that uncouple obesity from metabolic dysfunction.
Collapse
Affiliation(s)
- Fangluo Chen
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Leandro M Velez
- Center for Epigenetics and Metabolism, University of California, IrvineIrvineUnited States
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marcus M Seldin
- Center for Epigenetics and Metabolism, University of California, IrvineIrvineUnited States
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
3
|
Chen F, Saqib M, Terrillion CE, Miranda C, Sarver DC, Scafidi J, Wong GW. Role of CTRP14/C1QL1 in motor coordination and learning across the lifespan. Physiol Behav 2025; 291:114799. [PMID: 39761721 PMCID: PMC11788040 DOI: 10.1016/j.physbeh.2025.114799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
C1q/TNF-related protein 14 (CTRP14), also known as C1q-like 1 (C1QL1), is a synaptic protein predominantly expressed in the brain. It plays a critical role in the formation and maintenance of the climbing fiber-Purkinje cell synapses, ensuring that only one single winning climbing fiber from the inferior olivary neuron synapses with the proximal dendrites of Purkinje cells during the early postnatal period. Loss of CTRP14/C1QL1 results in incomplete elimination of supernumerary climbing fibers, leading to multiple persistent climbing fibers synapsing with the Purkinje cells. While this deficit impairs oculomotor learning in adult mice, the impact of CTRP14 deficiency on motor function throughout adulthood has not been examined. Here, we conduct behavioral tests on a constitutive Ctrp14 knockout (KO) mouse model to determine whether CTRP14 is required for motor learning and function in mice across the lifespan. We show that CTRP14 deficiency does not affect grip strength, nor sprint and endurance running, in young and old mice of either sex. We performed accelerated rotarod tests on mice at 6, 12, and 18 months old to assess motor coordination and learning. No significant differences were observed between WT and Ctrp14-KO mice of either sex across the lifespan. Lastly, we performed complex running wheel tests to detect latent motor deficits and found that aged Ctrp14-KO mice have intact motor skills. Despite some limitations of the study, our data suggest that CTRP14 is dispensable for gross motor skills, coordination, and learning throughout adulthood based on the specific tests performed.
Collapse
Affiliation(s)
- Fangluo Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chantelle E Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chloe Miranda
- Loyola University Maryland, Baltimore, MD 21205, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joseph Scafidi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Michael V. Johnston Center for Developmental Neuroscience, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
Chen F, Sarver DC, Saqib M, Velez LM, Aja S, Seldin MM, Wong GW. Loss of CTRP10 results in female obesity with preserved metabolic health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.01.565163. [PMID: 37961647 PMCID: PMC10635050 DOI: 10.1101/2023.11.01.565163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Obesity is a major risk factor for type 2 diabetes, dyslipidemia, cardiovascular disease, and hypertension. Intriguingly, there is a subset of metabolically healthy obese (MHO) individuals who are seemingly able to maintain a healthy metabolic profile free of metabolic syndrome. The molecular underpinnings of MHO, however, are not well understood. Here, we report that CTRP10/C1QL2-deficient mice represent a unique female model of MHO. CTRP10 modulates weight gain in a striking and sexually dimorphic manner. Female, but not male, mice lacking CTRP10 develop obesity with age on a low-fat diet while maintaining an otherwise healthy metabolic profile. When fed an obesogenic diet, female Ctrp10 knockout (KO) mice show rapid weight gain. Despite pronounced obesity, Ctrp10 KO female mice do not develop steatosis, dyslipidemia, glucose intolerance, insulin resistance, oxidative stress, or low-grade inflammation. Obesity is largely uncoupled from metabolic dysregulation in female KO mice. Multi-tissue transcriptomic analyses highlighted gene expression changes and pathways associated with insulin-sensitive obesity. Transcriptional correlation of the differentially expressed gene (DEG) orthologous in humans also shows sex differences in gene connectivity within and across metabolic tissues, underscoring the conserved sex-dependent function of CTRP10. Collectively, our findings suggest that CTRP10 negatively regulates body weight in females, and that loss of CTRP10 results in benign obesity with largely preserved insulin sensitivity and metabolic health. This female MHO mouse model is valuable for understanding sex-biased mechanisms that uncouple obesity from metabolic dysfunction.
Collapse
Affiliation(s)
- Fangluo Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dylan C. Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leandro M Velez
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcus M. Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - G. William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Zhu Z, Niu Q, Tang S, Jiang Y. Association between circulating CTRP9 levels and coronary artery disease: a systematic review and meta-analysis. PeerJ 2024; 12:e18488. [PMID: 39575169 PMCID: PMC11580665 DOI: 10.7717/peerj.18488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Background C1q tumor necrosis factor (TNF) related proteins 9 (CTRP9) is a novel adipocytokine that has been shown to have a cardioprotective effect in coronary artery disease (CAD). However, there are conflicting results on circulating levels of CTRP9 in patients with and without CAD. This meta-analysis was conducted to investigate the association between circulating CTRP9 levels and CAD. Objective The aim of this meta-analysis was to re-examine the relationship between circulating CTRP9 levels and CAD. Methods We searched PubMed, Web of Science, Embase, Cochrane Library, CNKI, VIP, Wanfang Data, and CBM for relevant studies up to October 2023, and 193 articles were identified. After reading the title, abstract and full text, a total of 25 articles were included in this meta-analysis. A prespecified protocol registered at INPLASY was followed (INPLASY202450066). Due to the high heterogeneity, we performed subgroup analyses and meta-regression based on patient characteristics, complications, clinical biochemical indicators, coronary artery lesion, and CAD classification. Publication bias was assessed using Egger's linear regression tests, Begg's rank correlation tests, and funnel plots. Results The results showed that the patient with CAD had significantly lower circulating CTRP9 levels than the control group (Z = 3.26, P = 0.001). Subgroup analysis and meta-regression findings demonstrated that observed heterogeneity could be attributed to population distribution. Patient characteristics (year of publication, patients' age, and BMI), complications (diabetes and type 2 diabetes mellitus (T2DM)), clinical biochemical indicators, coronary artery lesion (stability of coronary atherosclerotic plaque, and the number of diseased coronary vessels), and classification of CAD were not identified as source of heterogeneity. Conclusions The meta-analysis confirmed that circulating CTRP9 levels in CAD patients are significantly lower than those in patients without CAD. The association may be modified by the population distribution.
Collapse
Affiliation(s)
- Ziyi Zhu
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Qingsheng Niu
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
- Department of Emergency Medicine, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Emergency Medicine, Disaster Medical Center, West China Hospital of Sichuan University, Chengdu, China
| | - Shiyuan Tang
- Department of Emergency Medicine, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Emergency Medicine, Disaster Medical Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yaowen Jiang
- Department of Emergency Medicine, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Emergency Medicine, Disaster Medical Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Guan H, Xu H, Yan B, Xiang A, Chen X, Yu Q, Xu L. CTRP9: An Anti-Atherosclerotic Factor in ApoE Knockout Mice through Oxidative Stress Inhibition. FRONT BIOSCI-LANDMRK 2024; 29:339. [PMID: 39344333 DOI: 10.31083/j.fbl2909339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND C1q/tumor necrosis factor-related protein-9 (CTRP9) is critically involved in the pathophysiology of metabolic and cardiovascular disorders. This investigation aimed to clarify the mechanism underlying the role of CTRP9 in atherosclerosis in apolipoprotein E (ApoE) knockout (KO) mice. METHODS ApoE KO mice were fed a Western diet and injected with a virus which resulted in CTRP9 overexpression or knockdown for 12 weeks. The plasma lipid levels and atherosclerotic plaque areas were measured after the mice were euthanized. Aortas were isolated, and RNA sequencing was performed to identify the differentially expressed genes and related signaling pathways. Finally, plasma oxidative stress factors were measured to demonstrate the reliability of the RNA sequencing results. RESULTS The plasma lipid levels in the CTRP9 overexpression group did not significantly differ from those in the green fluorescence protein (GFP) group. Markablely, CTRP9 overexpression inhibited atherosclerotic plaque formation in ApoE KO mice, whereas CTRP9 knockdown promoted plaque formation. RNA sequencing analysis identified 3485 differentially expressed genes that were prominently enriched across 55 signaling pathways. Additionally, plasma oxidative stress factors were significantly reduced after CTRP9 overexpression, whereas these factors were increased after CTRP9 knockdown, which was consistent with the results of the RNA sequencing analysis. CONCLUSIONS These findings demonstrated that CTRP9 alleviated inflammation and cholesterol metabolism, which reduced oxidative stress in an atherosclerotic animal model. These beneficial effects may mediate the suppression of lesion development in the aorta.
Collapse
Affiliation(s)
- Hua Guan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Hao Xu
- Department of Anatomy, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Bin Yan
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Aoqi Xiang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Xiaochang Chen
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Qi Yu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Lixian Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| |
Collapse
|
7
|
Lei S, Li X, Zuo A, Ruan S, Guo Y. CTRP9 alleviates diet induced obesity through increasing lipolysis mediated by enhancing autophagy-initiation complex formation. J Nutr Biochem 2024; 131:109694. [PMID: 38906337 DOI: 10.1016/j.jnutbio.2024.109694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Recently, emerging evidence has suggested that obesity become a prevalent health threat worldwide. Reportedly, CTRP9 can ameliorate HFD induced obesity. However, the molecular mechanism underlying the role of CTRP9 in obesity remains elusive. In this study, we reported its major function in the regulation of lipolysis. First, we found that the expression of CTRP9 was decreased in mature adipocytes and white adipose tissue of obese mice. Then, we showed that overexpression adipose tissue CTRP9 alleviated diet-induced obesity and adipocytes hypertrophy, improved glucose intolerance and raised energy expenditure. Moreover, CTRP9 increased the lipolysis in vitro and vivo. Additionally, we determined that CTRP9 enhanced autophagy flux in adipocytes. Intriguingly, knock down Beclin1 by SiRNA abolished the effect of CTRP9 on lipolysis. Mechanically, CTRP9 enhanced the expression of SNX26. We demonstrated that SNX26 was a component of the ATG14L-Beclin1-VPS34 complex and enhanced the assembly of the autophagy-initiation complex. Collectively, our results suggested that CTRP9 alleviated diet induced obesity through enhancing lipolysis mediated by autophagy-initiation complex formation.
Collapse
Affiliation(s)
- Shengyun Lei
- Department of General Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012,Jinan,Shandong, China
| | - Xuehui Li
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China
| | - Anju Zuo
- Department of General Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012,Jinan,Shandong, China
| | - Shiyan Ruan
- Department of General Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012,Jinan,Shandong, China
| | - Yuan Guo
- Department of General Medicine, Qilu Hospital of Shandong University,107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012,Jinan,Shandong, China.
| |
Collapse
|
8
|
Cope H, Elsborg J, Demharter S, McDonald JT, Wernecke C, Parthasarathy H, Unadkat H, Chatrathi M, Claudio J, Reinsch S, Avci P, Zwart SR, Smith SM, Heer M, Muratani M, Meydan C, Overbey E, Kim J, Chin CR, Park J, Schisler JC, Mason CE, Szewczyk NJ, Willis CRG, Salam A, Beheshti A. Transcriptomics analysis reveals molecular alterations underpinning spaceflight dermatology. COMMUNICATIONS MEDICINE 2024; 4:106. [PMID: 38862781 PMCID: PMC11166967 DOI: 10.1038/s43856-024-00532-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/23/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Spaceflight poses a unique set of challenges to humans and the hostile spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. METHODS To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on space flown murine transcriptomic datasets focused on the skin, biochemical profiles of 50 NASA astronauts and human transcriptomic datasets generated from blood and hair samples of JAXA astronauts, as well as blood samples obtained from the NASA Twins Study, and skin and blood samples from the first civilian commercial mission, Inspiration4. RESULTS Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation are identified as potential drivers for skin health risks during spaceflight. Additionally, a machine learning model is utilized to determine gene pairings associated with spaceflight response in the skin. While we identified spaceflight-induced dysregulation, such as alterations in genes associated with skin barrier function and collagen formation, our results also highlight the remarkable ability for organisms to re-adapt back to Earth via post-flight re-tuning of gene expression. CONCLUSION Our findings can guide future research on developing countermeasures for mitigating spaceflight-associated skin damage.
Collapse
Affiliation(s)
- Henry Cope
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
| | - Jonas Elsborg
- Department of Energy Conversion and Storage, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Abzu, Copenhagen, 2150, Denmark
| | | | - J Tyson McDonald
- Department of Radiation Medicine, School of Medicine, Georgetown University, Washington D.C., WA, 20057, USA
| | - Chiara Wernecke
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Department of Aerospace and Geodesy, TUM School of Engineering and Design, Technical University of Munich, Munich, Germany
| | - Hari Parthasarathy
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- College of Engineering and Haas School of Business, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Hriday Unadkat
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- School of Engineering and Applied Science, Princeton University, Princeton, NJ, 08540, USA
| | - Mira Chatrathi
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- College of Letters and Science, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jennifer Claudio
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA
| | - Sigrid Reinsch
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA
| | - Pinar Avci
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337, Munich, Germany
| | - Sara R Zwart
- University of Texas Medical Branch, Galveston, TX, USA
| | - Scott M Smith
- Biomedical Research and Environmental Sciences Division, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, 77058, USA
| | - Martina Heer
- IU International University of Applied Sciences, Erfurt and University of Bonn, Bonn, Germany
| | - Masafumi Muratani
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, 305-8575, Japan
- Department of Genome Biology, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Cem Meydan
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Eliah Overbey
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Jangkeun Kim
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Christopher R Chin
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Jiwoon Park
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, USA
| | - Jonathan C Schisler
- McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, USA
| | - Nathaniel J Szewczyk
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Craig R G Willis
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Amr Salam
- St John's Institute of Dermatology, King's College London, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Afshin Beheshti
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
9
|
Chen F, Sarver DC, Saqib M, Zhou M, Aja S, Seldin MM, Wong GW. CTRP13 ablation improves systemic glucose and lipid metabolism. Mol Metab 2023; 78:101824. [PMID: 37844630 PMCID: PMC10598410 DOI: 10.1016/j.molmet.2023.101824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/30/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVE Tissue crosstalk mediated by secreted hormones underlies the integrative control of metabolism. We previously showed that CTRP13/C1QL3, a secreted protein of the C1q family, can improve glucose metabolism and insulin action in vitro and reduce food intake and body weight in mice when centrally delivered. A role for CTRP13 in regulating insulin secretion in isolated islets has also been demonstrated. It remains unclear, however, whether the effects of CTRP13 on cultured cells and in mice reflect the physiological function of the protein. Here, we use a loss-of-function mouse model to address whether CTRP13 is required for metabolic homeostasis. METHODS WT and Ctrp13 knockout (KO) mice fed a standard chow or a high-fat diet were subjected to comprehensive metabolic phenotyping. Transcriptomic analyses were carried out on visceral and subcutaneous fat, liver, and skeletal muscle to identify pathways altered by CTRP13 deficiency. RNA-seq data was further integrated with the Metabolic Syndrome in Man (METSIM) cohort data. Adjusted regression analysis was used to demonstrate that genetic variation of CTRP13 expression accounts for a significant proportion of variance between differentially expressed genes (DEGs) in adipose tissue and metabolic traits in humans. RESULTS Contrary to expectation, chow-fed Ctrp13-KO male mice had elevated physical activity, lower body weight, and improved lipid handling. On a high-fat diet (HFD), Ctrp13-KO mice of either sex were consistently more active and leaner. Loss of CTRP13 reduced hepatic glucose output and improved glucose tolerance, insulin sensitivity, and triglyceride clearance, though with notable sex differences. Consistent with the lean phenotype, transcriptomic analyses revealed a lower inflammatory profile in visceral fat and liver. Reduced hepatic steatosis was correlated with the suppression of lipid synthesis and enhanced lipid catabolism gene expression. Visceral fat had the largest number of DEGs and mediation analyses on the human orthologs of the DEGs suggested the potential causal contribution of CTRP13 to human metabolic syndrome. CONCLUSIONS Our results suggest that CTRP13 is a negative metabolic regulator, and its deficiency improves systemic metabolic profiles. Our data also suggest the reduction in circulating human CTRP13 levels seen in obesity and diabetes may reflect a compensatory physiologic response to counteract insulin resistance.
Collapse
Affiliation(s)
- Fangluo Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mingqi Zhou
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Tan Y, Feng P, Feng L, Shi L, Song Y, Yang J, Duan W, Gao E, Liu J, Yi D, Zhang B, Sun Y, Yi W. Low-dose exercise protects the heart against established myocardial infarction via IGF-1-upregulated CTRP9 in male mice. MedComm (Beijing) 2023; 4:e411. [PMID: 38020715 PMCID: PMC10674078 DOI: 10.1002/mco2.411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/13/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Regular exercise is recommended as an important component of therapy for cardiovascular diseases in clinical practice. However, there are still major challenges in prescribing an optimized exercise regimen to individual patients with established cardiac disease. Here, we tested the effects of different exercise doses on cardiac function in mice with established myocardial infarction (MI). Exercise was introduced to mice with MI after 4 weeks of surgery. Low-dose exercise (15 min/day for 8 weeks) improved mortality and cardiac function by increasing 44.39% of ejection fractions while inhibiting fibrosis by decreasing 37.74% of distant region. Unlike higher doses of exercise, low-dose exercise consecutively upregulated cardiac expression of C1q complement/tumor necrosis factor-associated protein 9 (CTRP9) during exercise (>1.5-fold). Cardiac-specific knockdown of CTRP9 abolished the protective effects of low-dose exercise against established MI, while cardiac-specific overexpression of CTRP9 protected the heart against established MI. Mechanistically, low-dose exercise upregulated the transcription factor nuclear receptor subfamily 2 group F member 2 by increasing circulating insulin-like growth factor 1 (IGF-1), therefore, upregulating cardiac CTRP9 expression. These results suggest that low-dose exercise protects the heart against established MI via IGF-1-upregulated CTRP9 and may contribute to the development of optimized exercise prescriptions for patients with MI.
Collapse
Affiliation(s)
- Yanzhen Tan
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Pan Feng
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Lele Feng
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Lei Shi
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Yujie Song
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Jian Yang
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Weixun Duan
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Erhe Gao
- Center for Translational MedicineLewis Katz School of Medicine at Temple UniversityPhiladelphiaPennsylvaniaUSA
| | - Jincheng Liu
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Dinghua Yi
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Bing Zhang
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Yang Sun
- Department of General MedicineXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Wei Yi
- Department of Cardiovascular SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
11
|
Song R, Hu W, Cheng R, Zhao Y, Qin W, Li X, Zhu Y, Gan L, Liu J. Association Between Circulating Levels of C1q/TNF-Related Protein-9 and Type 2 Diabetes Mellitus: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab 2023; 108:2728-2738. [PMID: 37029975 PMCID: PMC10505529 DOI: 10.1210/clinem/dgad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/10/2023] [Accepted: 03/22/2023] [Indexed: 04/09/2023]
Abstract
CONTEXT According to growing research, C1q/TNF-Related Protein-9 (CTRP9) appears to be linked to type 2 diabetes mellitus (T2DM). But the literature on circulating levels of CTRP9 in patients with T2DM has been contradictory. OBJECTIVE This is a systematic review and meta-analysis to reassess the circulating level of CTRP9 in patients with T2DM, with and without complications. METHODS Relevant studies published until October 31, 2021, were identified from the PubMed, Embase, Web of Science, Cochrane Library, WanFang, CNKI, VIP, and CBM databases. Participants with age ≥18 years with clinically diagnosed T2DM were included. Sex and diabetes complications were not restricted. The data were extracted by 2 reviewers independently using a standard data collection form. RESULTS Analysis demonstrated significantly lower circulating levels of CTRP9 in patients with T2DM than in patients without diabetes (standardized mean difference [SMD] = -1.36; 95% CI -1.78 to -0.93; P < .001), I2 = 97.5%, P < .001). Furthermore, the circulating level of CTRP9 in patients with T2DM-related complications was lower than that in patients with T2DM without complications, regardless of macrovascular complications or microvascular complications (SMD = -1.062; 95% CI -1.466 to -0.658; P < .001, I2 = 91.3%, P < .001). Subgroup analyses revealed that factors such as body mass index, T2DM duration, and fasting blood glucose were the sources of heterogeneity (P = .047, P = .034, and P = .07, respectively). CONCLUSION The present systematic review and meta-analysis found CTRP9 levels were lower in T2DM patients with or without complications. However, since this was a meta-analysis of most observational studies, these findings still need to be verified by further studies with a large sample size.
Collapse
Affiliation(s)
- Rui Song
- The Second Clinical Medical College of Shanxi Medical University, Taiyuan 030000, China
| | - Weiting Hu
- The Second Clinical Medical College of Shanxi Medical University, Taiyuan 030000, China
| | - Rui Cheng
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Yibo Zhao
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Weiwei Qin
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Xing Li
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Yikun Zhu
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Lu Gan
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Jing Liu
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| |
Collapse
|
12
|
Tang S, Li R, Ma W, Lian L, Gao J, Cao Y, Gan L. Cardiac-to-adipose axis in metabolic homeostasis and diseases: special instructions from the heart. Cell Biosci 2023; 13:161. [PMID: 37667400 PMCID: PMC10476430 DOI: 10.1186/s13578-023-01097-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/30/2023] [Indexed: 09/06/2023] Open
Abstract
Adipose tissue is essential for maintaining systemic metabolic homeostasis through traditional metabolic regulation, endocrine crosstalk, and extracellular vesicle production. Adipose dysfunction is a risk factor for cardiovascular diseases. The heart is a traditional pump organ. However, it has recently been recognized to coordinate interorgan cross-talk by providing peripheral signals known as cardiokines. These molecules include specific peptides, proteins, microRNAs and novel extracellular vesicle-carried cargoes. Current studies have shown that generalized cardiokine-mediated adipose regulation affects systemic metabolism. Cardiokines regulate lipolysis, adipogenesis, energy expenditure, thermogenesis during cold exposure and adipokine production. Moreover, cardiokines participate in pathological processes such as obesity, diabetes and ischemic heart injury. The underlying mechanisms of the cardiac-to-adipose axis mediated by cardiokines will be further discussed to provide potential therapeutic targets for metabolic diseases and support a new perspective on the need to correct adipose dysfunction after ischemic heart injury.
Collapse
Affiliation(s)
- Songling Tang
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, West China School of Medicine, Sichuan University Chengdu, Chengdu, 610041, People's Republic of China
| | - Ruixin Li
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, West China School of Medicine, Sichuan University Chengdu, Chengdu, 610041, People's Republic of China
| | - Wen Ma
- Sichuan University-The Hong Kong Polytechnic University Institute for Disaster Management and Reconstruction, Chengdu, China
| | - Liu Lian
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, West China School of Medicine, Sichuan University Chengdu, Chengdu, 610041, People's Republic of China
| | - Jiuyu Gao
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, West China School of Medicine, Sichuan University Chengdu, Chengdu, 610041, People's Republic of China
| | - Yu Cao
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, West China School of Medicine, Sichuan University Chengdu, Chengdu, 610041, People's Republic of China.
- Sichuan University-The Hong Kong Polytechnic University Institute for Disaster Management and Reconstruction, Chengdu, China.
| | - Lu Gan
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, West China School of Medicine, Sichuan University Chengdu, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
13
|
Zhang H, Zhang-Sun ZY, Xue CX, Li XY, Ren J, Jiang YT, Liu T, Yao HR, Zhang J, Gou TT, Tian Y, Lei WR, Yang Y. CTRP family in diseases associated with inflammation and metabolism: molecular mechanisms and clinical implication. Acta Pharmacol Sin 2023; 44:710-725. [PMID: 36207402 PMCID: PMC10042840 DOI: 10.1038/s41401-022-00991-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/27/2022] [Indexed: 11/08/2022]
Abstract
C1q/tumor necrosis factor (TNF) related proteins (CTRPs) is a newly discovered adipokine family with conservative structure and ubiquitous distribution and is secreted by adipose tissues. Recently, CTRPs have attracted increasing attention due to the its wide-ranging effects upon inflammation and metabolism. To-date, 15 members of CTRPs (CTRP1-15) with the characteristic C1q domain have been characterized. Earlier in-depth phenotypic analyses of mouse models of CTRPs deficiency have also unveiled ample function of CTRPs in inflammation and metabolism. This review focuses on the rise of CTRPs, with a special emphasis on the latest discoveries with regards to the effects of the CTRP family on inflammation and metabolism as well as related diseases. We first introduced the structure of characteristic domain and polymerization of CTRPs to reveal its pleiotropic biological functions. Next, intimate association of CTRP family with inflammation and metabolism, as well as the involvement of CTRPs as nodes in complex molecular networks, were elaborated. With expanding membership of CTRP family, the information presented here provides new perspectives for therapeutic strategies to improve inflammatory and metabolic abnormalities.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Zi-Yin Zhang-Sun
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Cheng-Xu Xue
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Xi-Yang Li
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Yu-Ting Jiang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Tong Liu
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Hai-Rong Yao
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Juan Zhang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Tian-Tian Gou
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Ye Tian
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Wang-Rui Lei
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
14
|
Complement 1q/Tumor Necrosis Factor-Related Proteins (CTRPs): Structure, Receptors and Signaling. Biomedicines 2023; 11:biomedicines11020559. [PMID: 36831095 PMCID: PMC9952994 DOI: 10.3390/biomedicines11020559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Adiponectin and the other 15 members of the complement 1q (C1q)/tumor necrosis factor (TNF)-related protein (CTRP) family are secreted proteins composed of an N-terminal variable domain followed by a stalk region and a characteristic C-terminal trimerizing globular C1q (gC1q) domain originally identified in the subunits of the complement protein C1q. We performed a basic PubMed literature search for articles mentioning the various CTRPs or their receptors in the abstract or title. In this narrative review, we briefly summarize the biology of CTRPs and focus then on the structure, receptors and major signaling pathways of CTRPs. Analyses of CTRP knockout mice and CTRP transgenic mice gave overwhelming evidence for the relevance of the anti-inflammatory and insulin-sensitizing effects of CTRPs in autoimmune diseases, obesity, atherosclerosis and cardiac dysfunction. CTRPs form homo- and heterotypic trimers and oligomers which can have different activities. The receptors of some CTRPs are unknown and some receptors are redundantly targeted by several CTRPs. The way in which CTRPs activate their receptors to trigger downstream signaling pathways is largely unknown. CTRPs and their receptors are considered as promising therapeutic targets but their translational usage is still hampered by the limited knowledge of CTRP redundancy and CTRP signal transduction.
Collapse
|
15
|
Cope H, Elsborg J, Demharter S, Mcdonald JT, Wernecke C, Parthasarathy H, Unadkat H, Chatrathi M, Claudio J, Reinsch S, Zwart S, Smith S, Heer M, Muratani M, Meydan C, Overbey E, Kim J, Park J, Schisler J, Mason C, Szewczyk N, Willis C, Salam A, Beheshti A. More than a Feeling: Dermatological Changes Impacted by Spaceflight. RESEARCH SQUARE 2023:rs.3.rs-2367727. [PMID: 36798347 PMCID: PMC9934743 DOI: 10.21203/rs.3.rs-2367727/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Spaceflight poses a unique set of challenges to humans and the hostile Spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on spaceflown murine transcriptomic datasets focused on the skin, biomedical profiles from fifty NASA astronauts, and confirmation via transcriptomic data from JAXA astronauts, the NASA Twins Study, and the first civilian commercial mission, Inspiration4. Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation were determined to be involved with skin health risks during Spaceflight. Additionally, a machine learning model was utilized to determine key genes driving Spaceflight response in the skin. These results can be used for determining potential countermeasures to mitigate Spaceflight damage to the skin.
Collapse
|
16
|
Haustein R, Trogisch FA, Keles M, Hille S, Fuhrmann M, Weinzierl N, Hemanna S, Thackeray J, Dou Y, Zwadlo C, Froese N, Cordero J, Bengel F, Müller OJ, Bauersachs J, Dobreva G, Heineke J. C1q and Tumor Necrosis Factor Related Protein 9 Protects from Diabetic Cardiomyopathy by Alleviating Cardiac Insulin Resistance and Inflammation. Cells 2023; 12:cells12030443. [PMID: 36766785 PMCID: PMC9914367 DOI: 10.3390/cells12030443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
(1) Background: Diabetic cardiomyopathy is a major health problem worldwide. CTRP9, a secreted glycoprotein, is mainly expressed in cardiac endothelial cells and becomes downregulated in mouse models of diabetes mellitus; (2) Methods: In this study, we investigated the impact of CTRP9 on early stages of diabetic cardiomyopathy induced by 12 weeks of high-fat diet; (3) Results: While the lack of CTRP9 in knock-out mice aggravated insulin resistance and triggered diastolic left ventricular dysfunction, AAV9-mediated cardiac CTRP9 overexpression ameliorated cardiomyopathy under these conditions. At this early disease state upon high-fat diet, no fibrosis, no oxidative damage and no lipid deposition were identified in the myocardium of any of the experimental groups. Mechanistically, we found that CTRP9 is required for insulin-dependent signaling, cardiac glucose uptake in vivo and oxidative energy production in cardiomyocytes. Extensive RNA sequencing from myocardial tissue of CTRP9-overexpressing and knock-out as well as respective control mice revealed that CTRP9 acts as an anti-inflammatory mediator in the myocardium. Hence, CTRP9 knock-out exerted more, while CTRP9-overexpressing mice showed less leukocytes accumulation in the heart during high-fat diet; (4) Conclusions: In summary, endothelial-derived CTRP9 plays a prominent paracrine role to protect against diabetic cardiomyopathy and might constitute a therapeutic target.
Collapse
Affiliation(s)
- Ricarda Haustein
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Felix A. Trogisch
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Merve Keles
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Manuela Fuhrmann
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nina Weinzierl
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Shruthi Hemanna
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - James Thackeray
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Yanliang Dou
- Cardiovascular Genomics and Epigenomics, ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Carolin Zwadlo
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Natali Froese
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Julio Cordero
- Cardiovascular Genomics and Epigenomics, ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Frank Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Oliver J. Müller
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Gergana Dobreva
- Cardiovascular Genomics and Epigenomics, ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- DZHK, Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Joerg Heineke
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- DZHK, Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
17
|
Saeidi A, Nouri-Habashi A, Razi O, Ataeinosrat A, Rahmani H, Mollabashi SS, Bagherzadeh-Rahmani B, Aghdam SM, Khalajzadeh L, Al Kiyumi MH, Hackney AC, Laher I, Heinrich KM, Zouhal H. Astaxanthin Supplemented with High-Intensity Functional Training Decreases Adipokines Levels and Cardiovascular Risk Factors in Men with Obesity. Nutrients 2023; 15:nu15020286. [PMID: 36678157 PMCID: PMC9866205 DOI: 10.3390/nu15020286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
The aim of this study was to investigate the effects of 12 weeks of high-intensity training with astaxanthin supplementation on adipokine levels, insulin resistance and lipid profiles in males with obesity. Sixty-eight males with obesity were randomly stratified into four groups of seventeen subjects each: control group (CG), supplement group (SG), training group (TG), and training plus supplement group (TSG). Participants underwent 12 weeks of treatment with astaxanthin or placebo (20 mg/d capsule daily). The training protocol consisted of 36 sessions of high-intensity functional training (HIFT), 60 min/sessions, and three sessions/week. Metabolic profiles, body composition, anthropometrical measurements, cardio-respiratory indices and adipokine [Cq1/TNF-related protein 9 and 2 (CTRP9 and CTRP2) levels, and growth differentiation factors 8 and 15 (GDF8 and GDF15)] were measured. There were significant differences for all indicators between the groups (p < 0.05). Post-hoc analysis indicated that the levels of CTRP9, CTRP2, and GDF8 were different from CG (p < 0.05), although levels of GDF15 were similar to CG (p > 0.05). Levels of GDF8 were similar in the SG and TG groups (p > 0.05), with reductions of GDF15 levels in both training groups (p < 0.05). A total of 12 weeks of astaxanthin supplementation and exercise training decreased adipokines levels, body composition (weight, %fat), anthropometrical factors (BMI), and improved lipid and metabolic profiles. These benefits were greater for men with obesity in the TSG group.
Collapse
Affiliation(s)
- Ayoub Saeidi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj 66177-15175, Iran
| | - Akbar Nouri-Habashi
- Department of Exercise Physiology and Corrective Movements, Faculty of Sport Sciences, Urmia University, Urmia 57561-51818, Iran
- Correspondence: (A.N.-H.); (M.H.A.K.)
| | - Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sports Science, Razi University, Kermanshah 94Q5+6G3, Iran
| | - Ali Ataeinosrat
- Department of Physical Education and Sport Science, Science and Research Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | - Hiwa Rahmani
- Faculty of Physical Education and Sports Science, Alzahra University, Tehran 19938 93973, Iran
| | | | - Behnam Bagherzadeh-Rahmani
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar M3J+373, Iran
| | - Shahin Mahmoudi Aghdam
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | - Leila Khalajzadeh
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | - Maisa Hamed Al Kiyumi
- Department of Family Medicine and Public Health, Sultan Qaboos University Hospital, Muscat H5QC+36M, Oman
- Correspondence: (A.N.-H.); (M.H.A.K.)
| | - Anthony C. Hackney
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Katie M. Heinrich
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, KS 66506, USA
| | - Hassane Zouhal
- Laboratoire Mouvement, Sport, Santé, University of Rennes, M2S—EA 1274, 35000 Rennes, France
- Institut International des Sciences du Sport (2I2S), 35850 Irodouer, France
| |
Collapse
|
18
|
Fadaei R, Azadi SM, Laher I, Khazaie H. Increased Levels of ANGPTL3 and CTRP9 in Patients With Obstructive Sleep Apnea and Their Relation to Insulin Resistance and Lipid Metabolism and Markers of Endothelial Dysfunction. Lab Med 2023; 54:83-89. [PMID: 35976955 DOI: 10.1093/labmed/lmac073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE Obstructive sleep apnea (OSA) has a close relation with obesity and perturbation in adipokines and hepatokines, which are linked to OSA consequences such as insulin resistance, dyslipidemia, and endothelial dysfunction. This study aimed to assess the relation of C1q/TNF-related protein 9 (CTRP9) and angiopoietin-like protein 3 (ANGPTL3) with OSA and biochemical measurements. METHODS Serum levels of ANGPTL3, CTRP9, adiponectin, leptin, intercellular adhesion molecule 1 (ICAM-1), and vascular cell adhesion protein 1 (VCAM-1) were determined in 74 OSA patients and 27 controls using enzyme-linked immunosorbent assay kits. RESULTS Levels of ANGPTL3, CTRP9, leptin, ICAM-1, and VCAM-1 were increased in the patients compared to the controls, whereas adiponectin levels decreased. ANGPTL3 had a positive correlation with total cholesterol, triglyceride, low-density lipoprotein cholesterol, ICAM-1, and VCAM-1 and was inversely correlated with leptin. CTRP9 showed a positive correlation with body mass index, insulin resistance, ICAM-1, and VCAM-1. CONCLUSION The results indicated the relation of ANGLTP3 and CTRP9 with OSA and its complications, which suggested a possible role for these factors in the consequences of OSA.
Collapse
Affiliation(s)
- Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Samaneh Mohassel Azadi
- Department of Clinical Biochemistry, Faculty of Medicine Tehran University of Medical Sciences, Tehran, Iran
| | - Ismail Laher
- Faculty of Medicine, Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, Canada
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
19
|
Kon M, Tanimura Y. Responses of complement C1q/tumor necrosis factor-related proteins to acute aerobic exercise. Cytokine 2023; 161:156083. [PMID: 36356496 DOI: 10.1016/j.cyto.2022.156083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/30/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
Abstract
Aerobic exercise is an effective therapeutic strategy to manage metabolic disorders. However, the mechanisms of aerobic exercise-induced improvements in metabolic diseases are not completely understood. Complement C1q/tumor necrosis factor-related protein (CTRP) 1, CTRP3, CTRP5, and CTRP9 have important roles in improving metabolic disorders via the adenosine monophosphate-activated protein kinase signaling pathway. In this study, we investigated the effects of acute aerobic exercise on circulating CTRP1, CTRP3, CTRP5, and CTRP9 levels in human participants. Eight healthy males with an age of 20.4 ± 0.2 years, height 173.1 ± 1.7 cm, body mass 68.0 ± 1.8 kg, body mass index 22.7 ± 0.7 kg/m2, and maximal oxygen uptake (VO2max) 51.3 ± 2.5 mL/kg/min performed acute aerobic cycling exercise at 75 % of their VO2max for 30 min (data are mean ± standard error). Blood samples were obtained before; immediately after; and 30, 60, and 120 min after exercising. Serum concentrations of CTRP1, CTRP3, CTRP5, CTRP9, tumor necrosis factor-α (TNF-α), and insulin were measured. The CTRP1 concentration significantly increased immediately after exercising and remained elevated for up to 120 min (p < 0.01). The CTRP3 concentration significantly increased at 60 min after exercise (p < 0.05), and the increasing trend continued until at least 120 min after exercise (p < 0.01). The CTRP5, CTRP9, TNF-α, and insulin concentrations significantly increased immediately after exercise (p < 0.05, p < 0.01, p < 0.05, and p < 0.05, respectively) and decreased thereafter. A significant correlation was observed between the peak post-exercise concentrations of CTRP1 and TNF-α (p < 0.05); however, no correlation was observed between the peak post-exercise concentrations of CTRP3 and insulin. The results of this study indicate that acute aerobic exercise may enhance the secretion of CTRP1, CTRP3, CTRP5, and CTRP9 in healthy adults.
Collapse
Affiliation(s)
- Michihiro Kon
- Department of Health Care and Sports, Faculty of Human Life Design, Toyo University, 1-7-11, Akabanedai, Kita-ku, Tokyo 115-0053, Japan.
| | - Yuko Tanimura
- Department of Sports Research, Japan Institute of Sports Sciences, 3-15-1 Nishigaoka, Kita-ku, Tokyo 115-0056, Japan
| |
Collapse
|
20
|
Yang MM, Balmert LC, Marangoni RG, Carns M, Hinchcliff M, Korman BD, Varga J. Circulating CTRP9 Is Associated With Severity of Systemic Sclerosis-Associated Interstitial Lung Disease. Arthritis Care Res (Hoboken) 2023; 75:152-157. [PMID: 34251759 PMCID: PMC9233895 DOI: 10.1002/acr.24749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/09/2021] [Accepted: 07/08/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE While interstitial lung disease (ILD) is the leading cause of morbidity and mortality in systemic sclerosis (SSc), there remains a paucity of predictive markers to assess disease progression. We previously demonstrated that adipose tissue metabolism and adipokine homeostasis is dysregulated in SSc. The present study was undertaken to determine the association and predictive ability of the novel adipokine C1q/tumor necrosis factor-related protein 9 (CTRP9) for SSc-associated ILD. METHODS We performed a retrospective longitudinal study utilizing the Northwestern Scleroderma Program Patient Registry and Biorepository. Serum levels of CTRP9 were measured in 110 SSc patients at baseline, and demographic, clinical, and pulmonary function test data were collected in 12-month intervals to 48 months. Longitudinal trajectory of forced vital capacity percent predicted (FVC%) was used as a primary outcome measure. We utilized a mixed model to compare trajectories of lung function by CTRP9 groups and performed latent trajectory analysis to accommodate for heterogeneity. RESULTS In cross-sectional analysis, elevated circulating CTRP9 was associated with significantly lower FVC% at baseline (72% ± 17 versus 80% ± 18; P = 0.02) and 48 months (68 ± 19 versus 84 ± 18; P = 0.001). In mixed model analysis, high CTRP9 was associated with worse lung function but not with a different trajectory (P = 0.23). In contrast, low CTRP9 identified patients with stability of lung disease with reasonable accuracy (sensitivity 73%). Latent trajectory analysis confirmed the association of lower CTRP9 with higher FVC%. CONCLUSION Higher circulating CTRP9 associated with worse pulmonary function, while low CTRP9 identified patients with lung disease stability over time. These findings suggest that CTRP9 may be a potential biomarker in SSc-associated ILD.
Collapse
Affiliation(s)
| | - Lauren C Balmert
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Mary Carns
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | | | | |
Collapse
|
21
|
Lee SM, Lee JW, Kim I, Woo DC, Pack CG, Sung YH, Baek IJ, Jung CH, Kim YH, Ha CH. Angiogenic adipokine C1q-TNF-related protein 9 ameliorates myocardial infarction via histone deacetylase 7-mediated MEF2 activation. SCIENCE ADVANCES 2022; 8:eabq0898. [PMID: 36459558 PMCID: PMC10936044 DOI: 10.1126/sciadv.abq0898] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 10/18/2022] [Indexed: 06/17/2023]
Abstract
C1q/tumor necrosis factor-related protein 9 (CTRP9) is an adipokine and has high potential as a therapeutic target. However, the role of CTRP9 in cardiovascular disease pathogenesis remains unclear. We found CTRP9 to induce HDAC7 and p38 MAPK phosphorylation via tight regulation of AMPK in vascular endothelial cells, leading to angiogenesis through increased MEF2 activity. The expression of CTRP9 and atheroprotective MEF2 was decreased in plaque tissue of atherosclerotic patients and the ventricle of post-infarction mice. CTRP9 treatment inhibited the formation of atherosclerotic plaques in ApoE KO and CTRP9 KO mice. In addition, CTRP9 induced significant ischemic injury prevention in the post-MI mice. Clinically, serum CTRP9 levels were reduced in patients with MI compared with healthy controls. In summary, CTRP9 induces a vasoprotective response via the AMPK/HDAC7/p38 MAPK pathway in vascular endothelial cells, whereas its absence can contribute to atherosclerosis and MI. Hence, CTRP9 may represent a valuable therapeutic target and biomarker in cardiovascular diseases.
Collapse
Affiliation(s)
- Seung Min Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inki Kim
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Cheol Woo
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Young Hoon Sung
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - In-Jeoung Baek
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chang Hee Jung
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Hak Kim
- Cardiology Division, Asan Medical Center and University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Hoon Ha
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
The Impact of Obesity on C1q/TNF-Related Protein-9 Expression and Endothelial Function following Acute High-Intensity Interval Exercise vs. Continuous Moderate-Intensity Exercise. BIOLOGY 2022; 11:biology11111667. [DOI: 10.3390/biology11111667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
C1q-TNF-related protein-9 (CTRP9) increases endothelial nitric oxide synthase and reduces vasoconstrictors. There is limited information regarding exercise-mediated CTRP9 in obesity. The purpose of this study was to compare high-intensity interval exercise (HIIE) and continuous moderate-intensity exercise (CME) on the CTRP9 response and an indicator of endothelial function (FMD) in obese participants. Sixteen young male participants (9 obese and 7 normal-weight) participated in a counterbalanced and caloric equated experiment: HIIE (30 min, 4 intervals of 4 min at 80–90% of VO2 max with 3 min rest between intervals) and CME (38 min at 50–60% VO2 max). Serum CTRP9 and FMD were measured prior to, immediately following exercise, and 1 h and 2 h into recovery. CTRP9 was significantly increased immediately following acute HIIE and CME in both groups (p = 0.003). There was a greater CME-induced FMD response at 2 h into recovery in obese participants (p = 0.009). A positive correlation between CTRP9 and FMD percent change was observed in response to acute CME when combined with both obese and normal-weight participants (r = 0.589, p = 0.016). The novel results from this study provide a foundation for additional examination of the mechanisms of exercise-mediated CTRP9 on endothelial function in individuals with obesity.
Collapse
|
23
|
Fadaei R, Goodarzi G, Yarahmadi S, Allahyari P, Fallah S, Moradi N. Circulating Levels of C1q/TNF-Related Protein 3 (CTRP3) and CTRP9 in Gestational Diabetes and Their Association with Insulin Resistance and Inflammatory Cytokines. Lab Med 2022; 54:262-269. [PMID: 36219707 DOI: 10.1093/labmed/lmac096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Abstract
Objective
Gestational diabetes mellitus (GDM) is closely related to obesity, adipose tissue, and adipokines. Adiponectin-homologous adipokines with anti-inflammatory properties, including C1q/TNF-related protein 3 (CTRP3) and CTRP9, regulate glucose and lipid metabolism, which was measured in pregnant women with GDM with the aim to assess their circulating levels and their relation with inflammatory cytokines and other biochemical data.
Methods
Serum levels of CTRP3, CTRP9, adiponectin, tumor necrosis factor (TNF)-α, and interleukin (IL)-6 were measured in 43 subjects with GDM and 42 healthy controls by enzyme-linked immunosorbent assay.
Results
Serum levels of adiponectin and CTRP3 were lower in GDM subjects than in controls, whereas CTRP9, TNF-α, and IL-6 showed higher concentrations in subjects with GDM than in controls. In the subjects with GDM, there was a significant association of CTRP3 with homeostasis model assessment of insulin resistance (HOMA-IR), body mass index, and triglycerides, whereas CTRP9 is associated with TNF-α and HOMA-IR.
Conclusion
The differences in the assessed levels of CTRP3 and CTRP9 suggest a possible relation with the pathogenesis of GDM, in particular insulin resistance, which showed significant association with both adipokines.
Collapse
Affiliation(s)
- Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Golnoosh Goodarzi
- Department of Nutrition, Science and Research Branch, Islamic Azad University , Tehran , Iran
| | - Sahar Yarahmadi
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences , Tehran , Iran
| | - Pooneh Allahyari
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Islamic Azad University, Central Tehran Branch , Tehran , Iran
| | - Soudabeh Fallah
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences , Tehran , Iran
| | - Nariman Moradi
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences , Sanandaj , Iran
| |
Collapse
|
24
|
Wang SX, Yan JS, Chan YS. Advancements in MAFLD Modeling with Human Cell and Organoid Models. Int J Mol Sci 2022; 23:11850. [PMID: 36233151 PMCID: PMC9569457 DOI: 10.3390/ijms231911850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolic (dysfunction) associated fatty liver disease (MAFLD) is one of the most prevalent liver diseases and has no approved therapeutics. The high failure rates witnessed in late-phase MAFLD drug trials reflect the complexity of the disease, and how the disease develops and progresses remains to be fully understood. In vitro, human disease models play a pivotal role in mechanistic studies to unravel novel disease drivers and in drug testing studies to evaluate human-specific responses. This review focuses on MAFLD disease modeling using human cell and organoid models. The spectrum of patient-derived primary cells and immortalized cell lines employed to model various liver parenchymal and non-parenchymal cell types essential for MAFLD development and progression is discussed. Diverse forms of cell culture platforms utilized to recapitulate tissue-level pathophysiology in different stages of the disease are also reviewed.
Collapse
Affiliation(s)
- Shi-Xiang Wang
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
| | - Ji-Song Yan
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yun-Shen Chan
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
| |
Collapse
|
25
|
Sarver DC, Xu C, Aja S, Wong GW. CTRP14 inactivation alters physical activity and food intake response to fasting and refeeding. Am J Physiol Endocrinol Metab 2022; 322:E480-E493. [PMID: 35403439 PMCID: PMC9126218 DOI: 10.1152/ajpendo.00002.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Secreted proteins of the C1q/TNF-related protein (CTRP) family play diverse functions in different organ systems. In the brain, CTRP14/C1QL1 is required for the proper establishment and maintenance of synapses between climbing fibers and cerebellar Purkinje cells. Beyond the central nervous system, the function of CTRP14 is largely unknown. A recent genome-wide association study has implicated CTRP14/C1QL1 as a candidate gene associated with total body fat mass. Here, we explored the potential metabolic roles of CTRP14. We show that Ctrp14 expression in peripheral tissues is dynamically regulated by fasting-refeeding and high-fat feeding. In the chow-fed basal state, Ctrp14 deletion modestly reduces glucose tolerance in knockout (KO) male mice and affects physical activity in a sex- and nutritional state-dependent manner. In the ad libitum fed state, Ctrp14 KO male mice have lower physical activity. In contrast, female KO mice have increased physical activity in the fasted and refed states. In response to an obesogenic diet, CTRP14-deficient mice of either sex gained similar weight and are indistinguishable from wild-type littermates in body composition, lipid profiles, and insulin sensitivity. Ambulatory activity, however, is reduced in Ctrp14 KO male mice. Food intake is also reduced in Ctrp14 KO male mice in the refed period following food deprivation. Meal pattern analyses indicate that decreased caloric intake from fasting to refeeding is due, in part, to smaller meal size. We conclude that CTRP14 is largely dispensable for metabolic homeostasis, but highlight context-dependent and sexually dimorphic metabolic responses of Ctrp14 deletion affecting physical activity and ingestive behaviors.NEW & NOTEWORTHY CTRP14 is a secreted protein whose function in the peripheral tissues is largely unknown. We show that the expression of Ctrp14 in peripheral tissues is regulated by metabolic and nutritional state. We generated mice lacking CTRP14 and show that CTRP14 deficiency alters physical activity and food intake in response to fasting and refeeding. Our data has provided new and valuable information on the physiological function of CTRP14.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Gao J, Lu J, Qiu J, Sun D, Xu B, Wang Z, Lei T. CTRP4
is Associated with Coronary Artery Disease in Patients with type 2 diabetes. J Diabetes Investig 2022; 13:1723-1731. [PMID: 35598316 PMCID: PMC9533036 DOI: 10.1111/jdi.13842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/15/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Aims/Introduction To evaluate the correlation of circulating C1q tumor necrosis factor‐related protein 4 (CTRP4) with coronary artery disease (CAD) in type 2 diabetes mellitus patients. Methods A total of 240 individuals with type 2 diabetes mellitus were enrolled in our center between January 2020 and December 2020. They were assigned into two groups, including the CAD and non‐CAD groups, based on coronary angiography or computed tomography angiography findings. Serum CTRP4 levels were detected by an enzyme‐linked immunosorbent assay kit. The association of CTRP4 with CAD was determined by logistic regression analysis. The predictive value of CTRP4 for CAD was calculated by receiver operating characteristic curve analysis. Results Median serum CTRP4 amounts were markedly elevated in the CAD group in comparison with the non‐CAD group (10.37 vs 3.75 ng/mL, P < 0.01). Binary logistic regression showed that CTRP4 was associated with CAD and even the amount of coronary artery lesions (P < 0.05). In receiver operating characteristic curve analysis, the area under the receiver operating characteristic curve was greater for CTRP4 compared with HbA1c or CRP (0.87 vs 0.74, 0.87 vs 0.80, P < 0.01). The area under the curve for CTRP4 and glycated hemoglobin in combination was larger than that obtained for CTRP4 combined with CRP (0.91 vs 0.87, P < 0.01). According to the maximum Youden index criteria, the optimal cut‐off of CTRP4 was 5.42 ng/mL, which yielded a sensitivity of 84.4% and a specificity of 76.7% in predicting CAD in type 2 diabetes mellitus patients. Conclusions Serum CTRP4 levels are positively correlated with CAD occurrence and severity. Combining CTRP4 and glycated hemoglobin has a better predictive value for CAD in type 2 diabetes mellitus patients.
Collapse
Affiliation(s)
- Jie Gao
- Department of Endocrinology, Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Jun Lu
- Department of Endocrinology, Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Junhui Qiu
- Department of Endocrinology, Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- Shanghai Putuo Central School of Clinical Medicine Anhui Medical University
| | - Dusang Sun
- Department of Endocrinology, Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Bilin Xu
- Department of Endocrinology, Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Zhihua Wang
- Department of Cardiology, Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Tao Lei
- Department of Endocrinology, Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- Shanghai Putuo Central School of Clinical Medicine Anhui Medical University
| |
Collapse
|
27
|
Sarver DC, Xu C, Carreno D, Arking A, Terrillion CE, Aja S, Wong GW. CTRP11 contributes modestly to systemic metabolism and energy balance. FASEB J 2022; 36:e22347. [PMID: 35579659 PMCID: PMC9164276 DOI: 10.1096/fj.202200189rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/23/2022] [Accepted: 04/29/2022] [Indexed: 12/18/2022]
Abstract
C1q/TNF‐related proteins (CTRP1‐15) constitute a conserved group of secreted proteins of the C1q family with diverse functions. In vitro studies have shown that CTRP11/C1QL4 can inhibit adipogenesis, antagonize myoblast fusion, and promote testosterone synthesis and secretion. Whether CTRP11 is required for these processes in vivo remains unknown. Here, we show that knockout (KO) mice lacking CTRP11 have normal skeletal muscle mass and function, and testosterone level, suggesting that CTRP11 is dispensable for skeletal muscle development and testosterone production. We focused our analysis on whether this nutrient‐responsive secreted protein plays a role in controlling sugar and fat metabolism. At baseline when mice are fed a standard chow, CTRP11 deficiency affects metabolic parameters in a sexually dimorphic manner. Only Ctrp11‐KO female mice have significantly higher fasting serum ketones and reduced physical activity. In the refeeding phase following food withdrawal, Ctrp11‐KO female mice have reduced food intake and increased metabolic rate and energy expenditure, highlighting CTRP11’s role in fasting–refeeding response. When challenged with a high‐fat diet to induce obesity and metabolic dysfunction, CTRP11 deficiency modestly exacerbates obesity‐induced glucose intolerance, with more pronounced effects seen in Ctrp11‐KO male mice. Switching to a low‐fat diet after obesity induction results in greater fat loss in wild type relative to KO male mice, suggesting impaired response to obesity reversal and reduced metabolic flexibility in the absence of CTRP11. Collectively, our data provide genetic evidence for novel sex‐dependent metabolic regulation by CTRP11, but note the overall modest contribution of CTRP11 to systemic energy homeostasis.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dana Carreno
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexander Arking
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chantelle E Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci 2022; 79:272. [PMID: 35503385 PMCID: PMC11073100 DOI: 10.1007/s00018-022-04286-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/11/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022]
Abstract
Characterized by a surplus of whole-body adiposity, obesity is strongly associated with the prognosis of atherosclerosis, a hallmark of coronary artery disease (CAD) and the major contributor to cardiovascular disease (CVD) mortality. Adipose tissue serves a primary role as a lipid-storage organ, secreting cytokines known as adipokines that affect whole-body metabolism, inflammation, and endocrine functions. Emerging evidence suggests that adipokines can play important roles in atherosclerosis development, progression, as well as regression. Here, we review the versatile functions of various adipokines in atherosclerosis and divide these respective functions into three major groups: protective, deteriorative, and undefined. The protective adipokines represented here are adiponectin, fibroblast growth factor 21 (FGF-21), C1q tumor necrosis factor-related protein 9 (CTRP9), and progranulin, while the deteriorative adipokines listed include leptin, chemerin, resistin, Interleukin- 6 (IL-6), and more, with additional adipokines that have unclear roles denoted as undefined adipokines. Comprehensively categorizing adipokines in the context of atherosclerosis can help elucidate the various pathways involved and potentially pave novel therapeutic approaches to treat CVDs.
Collapse
Affiliation(s)
- Longhua Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Zunhan Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiaohui Ji
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Wenqian Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jinwen Luan
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Tarik Zahr
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Li Qiang
- Department of Pathology and Cellular Biology and Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA.
| |
Collapse
|
29
|
Guan H, Wang Y, Li X, Xiang A, Guo F, Fan J, Yu Q. C1q/Tumor Necrosis Factor-Related Protein 9: Basics and Therapeutic Potentials. Front Physiol 2022; 13:816218. [PMID: 35370782 PMCID: PMC8971810 DOI: 10.3389/fphys.2022.816218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/23/2022] [Indexed: 01/19/2023] Open
Abstract
C1q/tumor necrosis factor-related protein 9 (CTRP9) is a newly discovered adipokine that is the closest paralog of adiponectin. Proteolytic cleavage of CTRP9 leads to the release of the globular domain (gCTRP9), which serves as the major circulating subtype. After binding with adiponectin receptor 1 (AdipoR1) and N-cadherin, CTRP9 activates various signaling pathways to regulate glucose and lipid metabolism, vasodilation and cell differentiation. Throughout human development and adult life, CTRP9 controls many biological phenomena. simultaneously, abnormal gene or protein expression of CTRP9 is accompanied by a wide range of human pathological phenomena. In this review, we briefly introduce CTRP9 and its associated signaling pathways and physiological functions, which may be helpful in the understanding of the occurrence of diseases. Moreover, we summarize the broader research prospects of CTRP9 and advances in therapeutic intervention. In recent years, CTRP9 has attracted extensive attention due to its role in the pathogenesis of various diseases, providing further avenues for its exploitation as a potential biomarker or therapeutic target.
Collapse
Affiliation(s)
- Hua Guan
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Yanli Wang
- Department of Pathology, Xi’an Medical University, Xi’an, China
| | - Xiangyu Li
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Aoqi Xiang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Fengwei Guo
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jianglin Fan
- Department of Pathology, Xi’an Medical University, Xi’an, China
- Department of Molecular Pathology, Faculty of Medicine, Interdisciplinary Graduate School of Medical Sciences, University of Yamanashi, Chuo, Japan
- *Correspondence: Jianglin Fan,
| | - Qi Yu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- Qi Yu,
| |
Collapse
|
30
|
Abstract
CONTEXT Resistance exercise training has recently been considered as an effective type of training to increase energy metabolism and insulin sensitivity. However, mechanisms of the resistance training-induced improvements in energy metabolism and insulin sensitivity have not been fully understood. Zinc-α2-glycoprotein (ZAG), which is a novel adipokine, has beneficial effects on energy metabolism and insulin sensitivity. OBJECTIVE We investigated the effect of a single bout of resistance exercise on the ZAG concentration. METHODS Nine healthy men were enrolled. They performed a single bout of resistance exercise (bench press and leg press) consisting of 10 repetitions of five sets at 70% of maximum strength with 90-s rests in between sets. Blood samples were obtained before and after acute resistance exercise to measure the ZAG concentration. RESULTS The serum ZAG concentration significantly increased following acute resistance exercise. CONCLUSION This result suggests that a single bout of resistance exercise may enhance the ZAG concentration.
Collapse
Affiliation(s)
- Michihiro Kon
- School of International Liberal Studies, Chukyo University, Nagoya, Japan
- Department of Sports Sciences, Japan Institute of Sports Sciences, Tokyo, Japan
| | - Yasuhiro Suzuki
- Department of Sports Research, Japan Institute of Sports Sciences, Tokyo, Japan
| |
Collapse
|
31
|
Sarver DC, Xu C, Cheng Y, Terrillion CE, Wong GW. CTRP4 ablation impairs associative learning and memory. FASEB J 2021; 35:e21910. [PMID: 34610176 DOI: 10.1096/fj.202100733rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 11/11/2022]
Abstract
C1q/TNF-related protein (CTRP) family comprises fifteen highly conserved secretory proteins with diverse central and peripheral functions. In zebrafish, mouse, and human, CTRP4 is most highly expressed in the brain. We previously showed that CTRP4 is a metabolically responsive regulator of food intake and energy balance, and mice lacking CTRP4 exhibit sexually dimorphic changes in ingestive behaviors and systemic metabolism. Recent single-cell RNA sequencing also revealed Ctrp4/C1qtnf4 expression in diverse neuronal cell types across distinct anatomical brain regions, hinting at additional roles in the central nervous system not previously characterized. To uncover additional central functions of CTRP4, we subjected Ctrp4 knockout (KO) mice to a battery of behavioral tests. Relative to wild-type (WT) littermates, loss of CTRP4 does not alter exploratory, anxiety-, or depressive-like behaviors, motor function and balance, sensorimotor gating, novel object recognition, and spatial memory. While pain-sensing mechanisms in response to thermal stress and mild shock are intact, both male and female Ctrp4 KO mice have increased sensitivity to pain induced by higher-level shock, suggesting altered nociceptive function. Importantly, CTRP4 deficiency impairs hippocampal-dependent associative learning and memory as assessed by trace fear conditioning paradigm. This deficit is sex-dependent, affects only female mice, and is associated with altered expression of learning and memory genes (Arc, c-fos, and Pde4d) in the hippocampus and cortex. Altogether, our behavioral and gene expression analyses have uncovered novel aspects of the CTRP4 function and provided a physiological context to further investigate its mechanism of action in the central and peripheral nervous system.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yi Cheng
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chantelle E Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Cheng Y, Qi Y, Liu S, Di R, Shi Q, Li J, Pei C. C1q/TNF-related Protein 9 Inhibits High Glucose-Induced Oxidative Stress and Apoptosis in Retinal Pigment Epithelial Cells Through the Activation of AMPK/Nrf2 Signaling Pathway. Cell Transplant 2021; 29:963689720962052. [PMID: 33040597 PMCID: PMC7784607 DOI: 10.1177/0963689720962052] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the common complications of diabetes mellitus. C1q/TNF-related protein 9 (CTRP9) has been demonstrated to be associated with the progression of diabetes and relative complications. However, its role in DR and underlying action of mechanism are not yet well understood. In the present study, human retinal pigment epithelial ARPE-19 cells were cultured under high concentration of glucose to simulate hyperglycemia condition in vitro. Our results showed that the expression of CTRP9 was significantly decreased in high glucose (HG)–stimulated ARPE-19 cells. CTRP9 overexpression improved HG-caused reduction in cell viability of ARPE-19 cells. CTRP9 overexpression significantly attenuated HG-induced oxidative stress, as proved by decreased levels of reactive oxygen species and malondialdehyde, and increased superoxide dismutase activity. Moreover, CTRP9 also prevented apoptosis in ARPE-19 cells in response to HG stimulation with decreased caspse-3 activity and bax expression, as well as increased bcl-2 expression. In contrast, knockdown of CTRP9 aggravated HG-induced oxidative stress and apoptosis. Furthermore, CTRP9 significantly induced the activation of AMPK/Nrf2 pathway in HG-induced ARPE-19 cells. Notably, inhibiting AMPK or Nrf2 blocked the protective effect of CTRP9 on ARPE-19 cells exposed to HG stimulation. Taken together, our findings suggested a protective effect of CTRP9 on HG-induced ARPE-19 cells and a putative mechanism involving the activation of AMPK/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yuhong Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Qi
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Siwei Liu
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rong Di
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiang Shi
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiayu Li
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Cheng Pei
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
33
|
Wolf RM, Jaffe AE, Rodriguez S, Lei X, Sarver DC, Straub AT, Wong GW, Magge SN. Altered adipokines in obese adolescents: a cross-sectional and longitudinal analysis across the spectrum of glycemia. Am J Physiol Endocrinol Metab 2021; 320:E1044-E1052. [PMID: 33900848 PMCID: PMC8285597 DOI: 10.1152/ajpendo.00626.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obesity and type 2 diabetes are rapidly increasing in the adolescent population. We sought to determine whether adipokines, specifically leptin, C1q/TNF-related proteins 1 (CTRP1) and CTRP9, and the hepatokine fibroblast growth factor 21 (FGF21), are associated with obesity and hyperglycemia in a cohort of lean and obese adolescents, across the spectrum of glycemia. In an observational, longitudinal study of lean and obese adolescents, we measured fasting laboratory tests, oral glucose tolerance tests, and adipokines including leptin, CTRP1, CTRP9, and FGF21. Participants completed baseline and 2-year follow-up study visits and were categorized as lean (LC, lean control; n = 30), obese normoglycemic (ONG; n = 61), and obese hyperglycemic (OHG; n = 31) adolescents at baseline and lean (n = 8), ONG (n = 18), and OHG (n = 4) at follow-up. Groups were compared using ANOVA and regression analysis, and linear mixed effects modeling was used to test for differences in adipokine levels across baseline and follow-up visits. Results showed that at baseline, leptin was higher in all obese groups (P < 0.001) compared with LC. FGF21 was higher in OHG participants compared with LC (P < 0.001) and ONG (P < 0.001) and positively associated with fasting glucose (P < 0.001), fasting insulin (P < 0.001), Homeostasis Model Assessment-Insulin Resistance Index (HOMA-IR; P < 0.001), and hemoglobin A1c (HbA1c; P = 0.01). CTRP1 was higher in OHG compared with ONG (P = 0.03). CTRP9 was not associated with obesity or hyperglycemia in this pediatric cohort. At 2 years, leptin decreased in ONG (P = 0.003) and FGF21 increased in OHG (P = 0.02), relative to lean controls. Altered adipokine levels are associated with the inflammatory milieu in obese youth with and without hyperglycemia. In adolescence, the novel adipokine CTRP1 was elevated with hyperglycemia, whereas CTRP9 was unchanged in this cohort.NEW & NOTEWORTHY Leptin is higher in obese adolescents and FGF21 is higher in obese hyperglycemic adolescents. The novel adipokine CTRP1 is higher in obese hyperglycemic adolescents, whereas CTRP9 was unchanged in this adolescent cohort.
Collapse
Affiliation(s)
- Risa M Wolf
- Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew E Jaffe
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Human Genetics, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Susana Rodriguez
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biochemistry, Oklahoma State University, Stillwater, Oklahoma
- Department of Molecular Biology, Oklahoma State University, Stillwater, Oklahoma
| | - Dylan C Sarver
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexander T Straub
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- University of Maryland, College Park, Maryland
| | - G William Wong
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sheela N Magge
- Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
34
|
Jerobin J, Ramanjaneya M, Bettahi I, Parammal R, Siveen KS, Alkasem M, Aye M, Sathyapalan T, Skarulis M, Atkin SL, Abou-Samra AB. Regulation of circulating CTRP-2/CTRP-9 and GDF-8/GDF-15 by intralipids and insulin in healthy control and polycystic ovary syndrome women following chronic exercise training. Lipids Health Dis 2021; 20:34. [PMID: 33874963 PMCID: PMC8054421 DOI: 10.1186/s12944-021-01463-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 04/06/2021] [Indexed: 11/10/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is associated with obesity, diabetes, and insulin resistance. The circulating C1Q/TNF-related proteins (CTRP-2, CTRP-9) and growth differentiation factors (GDF-8, GDF-15) contribute to glucose and lipid homeostasis. The effects of intralipids and insulin infusion on CTRP-2, CTRP-9, GDF-8 and GDF-15 in PCOS and control subjects before and after chronic exercise training were examined. Methods Ten PCOS and nine healthy subjects were studied at baseline status and after moderate-intensity chronic exercise training (1 h exercise, 3 times per week, 8 weeks). All participants were infused with 1.5 mL/min of saline or intralipids (20%) for 5 h, and during the last 2 h of saline or intralipids infusion hyperinsulinemic-euglycemic clamp (HIEC) was performed. CTRP-2, CTRP-9, GDF-8 and GDF-15 levels were measured at 0, 3 and 5 h. Results Intralipids dramatically increased CTRP-2 levels in PCOS (P = 0.02) and control (P = 0.004) subjects, which was not affected by insulin infusion or by exercise. Intralipids alone had no effects on CTRP-9, GDF-8, or GDF-15. Insulin increased the levels of GDF-15 in control subjects (P = 0.05) during the saline study and in PCOS subjects (P = 0.04) during the intralipid infusion. Insulin suppressed CTRP9 levels during the intralipid study in both PCOS (P = 0.04) and control (P = 0.01) subjects. Exercise significantly reduced fasting GDF-8 levels in PCOS (P = 0.03) and control (P = 0.04) subjects; however, intralipids infusion after chronic exercise training increased GDF-8 levels in both PCOS (P = 0.003) and control (P = 0.05) subjects and insulin infusion during intralipid infusion reduced the rise of GDF-8 levels. Conclusion This study showed that exogenous lipids modulate CTRP-2, which might have a physiological role in lipid metabolism. Since chronic exercise training reduced fasting GDF-8 levels; GDF-8 might have a role in humoral adaptation to exercise. GDF-15 and CTRP-9 levels are responsive to insulin, and thus they may play a role in insulin responses.
Collapse
Affiliation(s)
- Jayakumar Jerobin
- Qatar Metabolic Institute, Department of Medicine and Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Manjunath Ramanjaneya
- Qatar Metabolic Institute, Department of Medicine and Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ilham Bettahi
- Qatar Metabolic Institute, Department of Medicine and Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Raihanath Parammal
- Qatar Metabolic Institute, Department of Medicine and Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Meis Alkasem
- Qatar Metabolic Institute, Department of Medicine and Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Myint Aye
- Department of Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, UK
| | - Thozhukat Sathyapalan
- Department of Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, UK
| | - Monica Skarulis
- Qatar Metabolic Institute, Department of Medicine and Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Abdul Badi Abou-Samra
- Qatar Metabolic Institute, Department of Medicine and Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
35
|
Li L, Aslam M, Siegler BH, Niemann B, Rohrbach S. Comparative Analysis of CTRP-Mediated Effects on Cardiomyocyte Glucose Metabolism: Cross Talk between AMPK and Akt Signaling Pathway. Cells 2021; 10:cells10040905. [PMID: 33919975 PMCID: PMC8070942 DOI: 10.3390/cells10040905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
C1q/tumor necrosis factor -alpha-related proteins (CTRPs) have been shown to mediate protective cardiovascular effects, but no data exists on their effects on glucose and fatty acid (FA) metabolism in cardiomyocytes. In the present study, adult rat cardiomyocytes and H9C2 cardiomyoblasts were stimulated with various recombinant CTRPs. Glucose or FA uptake, expression of genes involved in glucose or FA metabolism and the role of the AMP-activated protein kinase (AMPK) and Akt were investigated. Although most CTRPs induced an increase in phosphorylation of AMPK and Akt in cardiomyocytes, mainly CTRP2, 7, 9 and 13 induced GLUT1 and GLUT4 translocation and glucose uptake in cardiomyocytes, despite high structural similarities among CTRPs. AMPK inhibition reduced the CTRPs-mediated activation of Akt, while Akt inhibition did not impair AMPK activation. In addition, CTRP2, 7, 9 and 13 mediated strong effects on the expression of enzymes involved in glucose or FA metabolism. Loss of adiponectin receptor 1, which has been suggested to be involved in CTRP-induced signal transduction, abolished the effects of some but not all CTRPs on glucose metabolism. Targeting the AMPK signaling pathway via CTRPs may offer a therapeutic principle to restore glucose homeostasis by acting on glucose uptake independent of the Akt pathway.
Collapse
Affiliation(s)
- Ling Li
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.H.S.); (S.R.)
- Correspondence: ; Tel.: +49-641-99-47342
| | - Muhammad Aslam
- Experimental Cardiology, Department of Cardiology and Angiology, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Benedikt H. Siegler
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.H.S.); (S.R.)
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.H.S.); (S.R.)
| |
Collapse
|
36
|
周 鹏, 刘 丽, 高 卫. [Association between serum CTRP9 levels and diabetic retinopathy in patients with type 2 diabetes mellitus]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:459-463. [PMID: 33849840 PMCID: PMC8075780 DOI: 10.12122/j.issn.1673-4254.2021.03.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To investigate the relationship between serum C1q tumor necrosis factor-related protein 9 (CTRP9) level and the risk of diabetic retinopathy (DR) in patients with type 2 diabetes mellitus (T2DM). OBJECTIVE A total of 291 patients with T2DM underwent fundus examination, and their serum levels of CTRP9, insulin and adiponectin were measured using enzyme- linked immunosorbent assay. According to results of fundus examination, the patients were divided into DR group and non-DR (NDR) group, and logistic regression was used to analyze the relationship between serum CTRP9 levels and DR in T2DM patients. OBJECTIVE Compared with those in NDR group, the patients with DR showed significantly increased serum CTRP9 level (P < 0.001) and decreased serum adiponectin level (P < 0.001). Pearson correlation analysis showed that in patients with T2DM complicated by DR, serum CTRP9 levels had a significant positive correlation with DR stage (P < 0.05) and a negative correlation with serum adiponectin level (P < 0.001). Multivariate logistic regression analysis showed that with the increase of serum CTRP9 level, the risk of DR is significantly increased in patients with T2DM. OBJECTIVE In patients with T2DM complicated by DR, an increased serum CTRP9 level suggests a compensatory response to DR.
Collapse
Affiliation(s)
- 鹏鹏 周
- 南京中医药大学附属昆山市中医医院,江苏 昆山 215300Kunshan Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan 215300, China
- 南京中医药大学,江苏 南京 210029Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - 丽燕 刘
- 南京中医药大学附属昆山市中医医院,江苏 昆山 215300Kunshan Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan 215300, China
| | - 卫萍 高
- 南京中医药大学,江苏 南京 210029Nanjing University of Chinese Medicine, Nanjing 210029, China
| |
Collapse
|
37
|
Liu D, Gu G, Gan L, Yan W, Zhang Z, Yao P, Zhu D, Lau WB, Xie D, Wu S, Meng Z, Tsukuda J, Christopher T, Lopez B, Zhao J, Gao E, Koch W, Ma XL, Wang Y. Identification of a CTRP9 C-Terminal polypeptide capable of enhancing bone-derived mesenchymal stem cell cardioprotection through promoting angiogenic exosome production. Redox Biol 2021; 41:101929. [PMID: 33714738 PMCID: PMC7966869 DOI: 10.1016/j.redox.2021.101929] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/07/2021] [Accepted: 02/28/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Mesenchymal stem cell therapy improves ischemic heart failure via incompletely understood mechanisms. C1q-TNFα related protein-9 (CTRP9) is a novel anti-oxidative cardiokine capable of improving the local microenvironment and cell survival by its c-terminal active globular domain (gCTRP9). The current study attempted to: 1) identify active gCTRP9 c-terminal polypeptides with stem cell protective function; 2) determine whether a lead polypeptide may enable/enhance cortical bone-derived mesenchymal stem cell (CBSC) cardioprotection against post-myocardial infarction (post-MI) remodeling; and 3) define the responsible underlying cellular/molecular mechanisms. METHODS AND RESULTS Utilizing I-TASSER structure prediction and 3-D active site modeling, we cloned and purified 3 gCTRP9 fragments (CTRP9-237, CTRP9-277, and CTRP9-281). Their activation of cell salvage kinase was compared against gCTRP9. Among the three fragments, CTRP9-281 (a 45 residue-containing polypeptide) exerted comparable or greater ERK1/2 activation compared to gCTRP9. Treatment with CTRP9-281 or gCTRP9 significantly increased CBSC proliferation and migration, and attenuated oxidative stress-induced CBSC apoptosis. CTRP9-281 and gCTRP9 comparably upregulated SOD2 and SOD3 expression. However, CTRP9-281, not gCTRP9, upregulated FGF2 and VEGFA expression/secretion in an ERK1/2 dependent manner. Administration of gCTRP9 or CTRP9-281 alone attenuated post-MI cardiac dysfunction and improved CBSC retention in the infarcted heart in similar fashion. However, CTRP9-281 exerted greater synergistic effect with CBSC than gCTRP9 related to pro-angiogenic, anti-fibrotic, and anti-remodeling effects. Mechanistically, CTRP9-281 significantly increased SOD2-rich and VEGFA-rich exosome production by CBSC. Exosomes from CTRP9-281 treated CBSC significantly attenuated oxidative stress-induced cardiomyocyte apoptosis in vitro. An exosome generation inhibitor attenuated CTRP9-281 enhancement of CBSC cardioprotection in vivo. CONCLUSION We identified a CTRP9 polypeptide that upregulates SOD2/SOD3 expression and improves CBSC survival/retention, similar to gCTRP9. Moreover, CTRP9-281 stimulates VEGFA-rich exosome production by CBSC, exerting superior pro-angiogenic, anti-fibrotic, and cardioprotective actions.
Collapse
Affiliation(s)
- Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Guoqiang Gu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Wenjun Yan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Zhen Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Peng Yao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Di Zhu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Sisi Wu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Zhijun Meng
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Jumpei Tsukuda
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Theodore Christopher
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Bernard Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Walter Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA,Corresponding author. Department of Emergency Medicine and Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA,Corresponding author.
| |
Collapse
|
38
|
Lei S, Chen J, Song C, Li J, Zuo A, Xu D, Li T, Guo Y. CTRP9 alleviates foam cells apoptosis by enhancing cholesterol efflux. Mol Cell Endocrinol 2021; 522:111138. [PMID: 33352225 DOI: 10.1016/j.mce.2020.111138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022]
Abstract
The apoptosis of foam cells leads to instability of atherosclerotic plaques. This study was designed to explore the protective role of CTRP9 in foam cell apoptosis. In our experiment, CTRP9 alleviated foam cell apoptosis. Meanwhile, CTRP9 upregulated the expression of proteins important for cholesterol efflux, such as LXRα, CYP27A1, ABCG1 and ABCA1, and improved cholesterol efflux in foam cells. Moreover, CTRP9 inhibited Wnt3a and β-catenin expression and β-catenin nuclear translocation in foam cells. In addition, adenovirus overexpression of Wnt3a abolished the effect of CTRP9 on macrophage apoptosis. Mechanistically, the AMPK inhibitor abolished the effect of CTRP9 on foam cell apoptosis, and downregulation of AdipoR1 by siRNA abrogated the activation of AMPK and the effect of CTRP9 on foam cell apoptosis. We concluded that CTRP9 achieved these protective effects on foam cells through the AdipoR1/AMPK pathway.
Collapse
Affiliation(s)
- Shengyun Lei
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Jiying Chen
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Chengxiang Song
- Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Jun Li
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Anju Zuo
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Dan Xu
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Tingting Li
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China.
| | - Yuan Guo
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China.
| |
Collapse
|
39
|
Rohrbach S, Li L, Novoyatleva T, Niemann B, Knapp F, Molenda N, Schulz R. Impact of PCSK9 on CTRP9-Induced Metabolic Effects in Adult Rat Cardiomyocytes. Front Physiol 2021; 12:593862. [PMID: 33643060 PMCID: PMC7904879 DOI: 10.3389/fphys.2021.593862] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
The adipocytokine adiponectin and its structural homologs, the C1q/TNF-related proteins (CTRPs), increase insulin sensitivity, fatty acid oxidation and mitochondrial biogenesis. Adiponectin- and CTRP-induced signal transduction has been described to involve the adiponectin receptors and a number of co-receptors including the Low density lipoprotein receptor-related protein 1 (LRP1). LRP1 is another target of the proprotein convertase subtilisin/kexin-9 (PCSK9) in addition to the LDL-receptor (LDL-R). Here, we investigated the influence of PCSK9 on the metabolic effects of CTRP9, the CTRP with the highest homology to adiponectin. Knockdown of LRP1 in H9C2 cardiomyoblasts blunts the effects of CTRP9 on signal transduction and mitochondrial biogenesis, suggesting its involvement in CTRP9-induced cellular effects. Treatment of adult rat cardiomyocytes with recombinant PCSK9 but not knockdown of endogenous PCSK9 by siRNA results in a strong reduction in LRP1 protein expression and subsequently reduces the mitochondrial biogenic effect of CTRP9. PCSK9 treatment (24 h) blunts the effects of CTRP9-induced signaling cascade activation (AMP-dependent protein kinase, protein kinase B). In addition, the stimulating effects of CTRP9 on cardiomyocyte mitochondrial biogenesis and glucose metabolism (GLUT-4 translocation, glucose uptake) are largely blunted. Basal fatty acid (FA) uptake is strongly reduced by exogenous PCSK9, although protein expression of the PCSK9 target CD36, the key regulator of FA transport in cardiomyocytes, is not altered. In addition, only minor effects of PCSK9 were observed on CTRP9-induced FA uptake or the expression of genes involved in FA metabolism or uptake. Finally, this CTRP9-induced increase in CD36 expression occurs independent from LRP1 and LDL-R. In conclusion, PCSK9 treatment influences LRP1-mediated signaling pathways in cardiomyocytes. Thus, therapeutic PCSK9 inhibition may provide an additional benefit through stimulation of glucose metabolism and mitochondrial biogenesis in addition to the known lipid-lowering effects. This could be an important beneficial side effect in situations with impaired mitochondrial function and reduced metabolic flexibility thereby influencing cardiac function.
Collapse
Affiliation(s)
- Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Ling Li
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University Giessen, Giessen, Germany
| | - Fabienne Knapp
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Nicole Molenda
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
40
|
Lee J, Yoo JH, Kim HS, Cho YK, Lee YL, Lee WJ, Park JY, Jung CH. C1q/TNF-related protein-9 attenuates palmitic acid-induced endothelial cell senescence via increasing autophagy. Mol Cell Endocrinol 2021; 521:111114. [PMID: 33301838 DOI: 10.1016/j.mce.2020.111114] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/27/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Autophagy is an important process in the pathogenesis of atherosclerosis. C1q/tumor necrosis factor-related protein 9 (CTRP9) is the closest adiponectin paralog. CTRP9 has anti-aging and anti-atherogenic effects, but its roles in autophagy and endothelial senescence are currently unknown. This study aimed to evaluate whether CTRP9 prevents palmitic acid (PA)-induced endothelial senescence by promoting autophagy. After no treatment or pre-treatment of human umbilical vein endothelial cells with CTRP9 prior to PA treatment, the level of senescence was measured by senescence associated acidic β-galactosidase staining and the level of hyperphosphorylated pRB protein. Autophagy was evaluated by LC3 conversion and the level of p62/SQSTM1, a protein degraded during autophagy. Autophagosome-lysosome fusion was detected by fluorescence microscopy. Pre-treatment with CTRP9 attenuated PA-induced endothelial senescence. CTRP9 increased the conversion of LC3-I to LC3-II and decreased p62 levels in a time- and dose-dependent manner. Although both CTRP9 and PA treatment increased LC3 conversion, treatment with PA increased the expression level of p62 and decreased the fusion of autophagosomes and lysosomes, which represented decreased autophagic flux. However, pre-treatment with CTRP9 recovered the autophagic flux inhibited by PA. AMP-activated kinase (AMPK) activation was involved in LC3 conversion and decreased p62 levels induced by CTRP9. CTRP9 inhibits PA-induced endothelial senescence by recovering autophagy and autophagic flux through AMPK activation.
Collapse
Affiliation(s)
- Jiwoo Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jee Hee Yoo
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Hwi Seung Kim
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yun Kyung Cho
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Yoo La Lee
- Asan Institute of Life Science, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Je Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joong-Yeol Park
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Chang Hee Jung
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Jiang N, Zhou S, Wang G, Jiang N, Wang H, Zhao F. Diagnostic value and prognostic significance of CTRP9 combined with pentraxin-3 in acute coronary syndrome. Exp Ther Med 2021; 21:254. [PMID: 33603861 PMCID: PMC7851676 DOI: 10.3892/etm.2021.9685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to explore the diagnostic value and prognostic significance of C1q/tumor necrosis factor-related protein 9 (CTRP9) combined with pentraxin-3 (PTX-3) in acute coronary syndrome (ACS). A total of 137 patients with coronary heart disease and chest pain were included. Among them, seventy-nine patients with ACS were allocated into a study group and fifty-eight patients with non-cardiac chest pain (NCCP) were allocated into a control group. The serum CTRP9, PTX-3 levels were quantified by ELISA, and their correlation with other ACS-related indexes, diagnostic value for ACS and predictive significance for poor prognosis were analyzed. In addition, the risk factors of the poor prognosis of ACS patients were studied. CTRP9 was lowly expressed and PTX-3 was highly expressed in the serum of ACS patients. CTRP9 was negatively correlated with cardiac troponin I (cTnI), creatine kinase-MB (CK-MB) and high-sensitivity C-reactive protein (hs-CRP) (P<0.05), while PTX-3 was positively correlated with them (P<0.05). Combined detection of CTRP9 and PTX-3 was of high value in the diagnosis and prognosis of ACS patients. In addition, CTRP9 and PTX-3 were independent risk factors for the poor prognosis of ACS. Patients with ACS had lower CTRP9 expression and higher PTX-3 expression than those without ACS. Moreover, the combined detection of CTRP9 and PTX-3 can better evaluate the diagnosis and prognosis of ACS patients.
Collapse
Affiliation(s)
- Na Jiang
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Shulong Zhou
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Guanglei Wang
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Ningning Jiang
- Department of Ophthalmology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Huaixin Wang
- Department of Emergency, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Feng Zhao
- Department of Cardiology, Dongying Hospital of Traditional Chinese Medicine, Dongying, Shandong 257000, P.R. China
| |
Collapse
|
42
|
Sarver DC, Stewart AN, Rodriguez S, Little HC, Aja S, Wong GW. Loss of CTRP4 alters adiposity and food intake behaviors in obese mice. Am J Physiol Endocrinol Metab 2020; 319:E1084-E1100. [PMID: 33017221 PMCID: PMC7792665 DOI: 10.1152/ajpendo.00448.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Central and peripheral mechanisms are both required for proper control of energy homeostasis. Among circulating plasma proteins, C1q/TNF-related proteins (CTRPs) have recently emerged as important regulators of sugar and fat metabolism. CTRP4, expressed in brain and adipose tissue, is unique among the family members in having two tandem globular C1q domains. We previously showed that central administration of recombinant CTRP4 suppresses food intake, suggesting a central nervous system role in regulating ingestive physiology. Whether this effect is pharmacological or physiological remains unclear. We used a loss-of-function knockout (KO) mouse model to clarify the physiological role of CTRP4. Under basal conditions, CTRP4 deficiency increased serum cholesterol levels and impaired glucose tolerance in male but not female mice fed a control low-fat diet. When challenged with a high-fat diet, male and female KO mice responded differently to weight gain and had different food intake patterns. On an obesogenic diet, male KO mice had similar weight gain as wild-type littermates. When fed ad libitum, KO male mice had greater meal number, shorter intermeal interval, and reduced satiety ratio. Female KO mice, in contrast, had lower body weight and adiposity. In the refeeding period following food deprivation, female KO mice had significantly higher food intake due to longer meal duration and reduced satiety ratio. Collectively, our data provide genetic evidence for a sex-dependent physiological role of CTRP4 in modulating food intake patterns and systemic energy metabolism.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ashley N Stewart
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
43
|
Yamaguchi S, Shibata R, Ohashi K, Enomoto T, Ogawa H, Otaka N, Hiramatsu-Ito M, Masutomi T, Kawanishi H, Murohara T, Ouchi N. C1q/TNF-Related Protein 9 Promotes Revascularization in Response to Ischemia via an eNOS-Dependent Manner. Front Pharmacol 2020; 11:1313. [PMID: 32973529 PMCID: PMC7472599 DOI: 10.3389/fphar.2020.01313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/07/2020] [Indexed: 12/25/2022] Open
Abstract
Strategies to promote revascularization are valuable for ischemic cardiovascular disease. Although C1q/TNF-related protein (CTRP) 9 is an adiponectin paralog with protective properties against cardiometabolic disorders, the role of endogenous CTRP9 in endothelial function is largely unknown. This study aimed to investigate the effects of CTRP9 on revascularization processes and dissected the potential mechanisms. CTRP9-knockout (KO) and wild-type (WT) mice were subjected to unilateral hindlimb ischemic surgery. CTRP9-KO mice exhibited impaired blood flow recovery and decreased capillary density in the ischemic limb compared with WT mice. In both CTRP9-KO and WT mice, systemic delivery of an adenoviral vector expressing CTRP9 (Ad-CTRP9) accelerated blood flow recovery. Treatment with recombinant CTRP9 protein increased network formation and migration of cultured human umbilical vein endothelial cells (HUVECs). CTRP9 promoted the phosphorylation of AMP-activated kinase (AMPK), Akt, and endothelial nitric oxide synthase (eNOS) in HUVECs. CTRP9-KO mice also showed reduced phosphorylation levels of AMPK, Akt, and eNOS in the ischemic limbs compared with WT mice. Furthermore, blockade of AMPK or Akt signaling pathway reversed the CTRP9-stimulated eNOS phosphorylation in HUVECs. Treatment with the NOS inhibitor significantly reduced CTRP9-stimulated network formation and migration of HUVECs. Of note, Ad-CTRP9 had no effects on blood flow of the ischemic limb in eNOS-KO mice. These results indicated that CTRP9 promotes endothelial cell function and ischemia-induced revascularization through the eNOS-dependent mechanism, suggesting that CTRP9 represents a target molecule for treatment of ischemic vascular diseases.
Collapse
Affiliation(s)
- Shukuro Yamaguchi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Enomoto
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hayato Ogawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoya Otaka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mizuho Hiramatsu-Ito
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiro Masutomi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kawanishi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
44
|
Masoodian SM, Toolabi K, Omidifar A, Zabihi H, Rahimipour A, Shanaki M. Increased mRNA Expression of CTRP3 and CTRP9 in Adipose Tissue from Obese Women: Is it Linked to Obesity-Related Parameters and mRNA Expression of Inflammatory Cytokines? Rep Biochem Mol Biol 2020; 9:71-81. [PMID: 32821754 DOI: 10.29252/rbmb.9.1.71] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Obesity, a medical condition with impaired adipokine secretion and function, has a detrimental effect on insulin and glucose metabolism. CTRP3 and CTRP9 are adipokines with possible roles in energy homeostasis regulation. We sought to compare CTRP3, CTRP9, and inflammatory gene expression in subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) from obese women who underwent bariatric surgery and non-obese women as controls. Methods For this study, the investigators recruited 20 morbidly obese women (BMI> 35) who qualified for bariatric surgery and 20 normal-weight women (BMI< 25) who underwent elective surgeries. Real-time PCR was performed to investigate mRNA expression of CTRP3, CTRP9, and the inflammatory genes IL1-β, IL-6, MCP-1, and TNF-α in SAT and VAT from both obese patients and controls. Results We observed that CTRP3 mRNA levels were significantly greater in VAT from obese patients than from controls (P< 0.0003). Also, patient group had higher levels of CTRP9 that control group (P< 0.0026). Inflammatory cytokines were markedly increased in SAT of obese patients compared to controls (P< 0.05). In addition, our results revealed a positive correlation of CTRP9 with HOMA-IR and waist circumference in VAT and CTRP3 with IL-1β, MCP-1, and TNF-α in SAT. Conclusion Both CTRP3 and CTRP9 expression were significantly higher in VAT from obese patients than from controls, and CTRP3 expression positively correlated with inflammatory parameters. Our findings indicate that CTRP3 and CTRP9 might be important in regulating glucose metabolism and obesity-related conditions such as inflammation.
Collapse
Affiliation(s)
- Seyed Mohammad Masoodian
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Karamollah Toolabi
- Department Surgery, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Omidifar
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Zabihi
- Sina Hospital, Department of Surgery, school of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Rahimipour
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Shanaki
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Tan SY, Little HC, Sarver DC, Watkins PA, Wong GW. CTRP12 inhibits triglyceride synthesis and export in hepatocytes by suppressing HNF-4α and DGAT2 expression. FEBS Lett 2020; 594:3227-3239. [PMID: 32749667 DOI: 10.1002/1873-3468.13895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022]
Abstract
C1q/TNF-related protein 12 (CTRP12) is an antidiabetic adipokine whose circulating levels are reduced in obesity and diabetes. Although partial and complete loss-of-function mouse models suggest a role for CTRP12 in modulating lipid metabolism and adiposity, its effect on cellular lipid metabolism remains poorly defined. Here, we demonstrate a direct action of CTRP12 in regulating lipid synthesis and secretion. In hepatoma cells and primary mouse hepatocytes, CTRP12 treatment inhibits triglyceride synthesis by suppressing glycerophosphate acyltransferase (GPAT) and diacylglycerol acyltransferase (DGAT) expression. CTRP12 treatment also downregulates the expression of hepatocyte nuclear factor-4α (HNF-4α) and its target gene microsomal triglyceride transfer protein (MTTP), leading to reduced very-low-density lipoprotein (VLDL)-triglyceride export from hepatocytes. Consistent with the in vitro findings, overexpressing CTRP12 lowers fasting and postprandial serum triglyceride levels in mice. These results underscore the important function of CTRP12 in lipid metabolism in hepatocytes.
Collapse
Affiliation(s)
- Stefanie Y Tan
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Pfizer, 1 Portland St., Cambridge, MA, 02139, USA
| | - Hannah C Little
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dylan C Sarver
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul A Watkins
- Department of Neurology and Biological Chemistry, Johns Hopkins University School of Medicine, and Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - G William Wong
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
46
|
Tan SY, Lei X, Little HC, Rodriguez S, Sarver DC, Cao X, Wong GW. CTRP12 ablation differentially affects energy expenditure, body weight, and insulin sensitivity in male and female mice. Am J Physiol Endocrinol Metab 2020; 319:E146-E162. [PMID: 32421370 PMCID: PMC7468785 DOI: 10.1152/ajpendo.00533.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Secreted hormones facilitate tissue cross talk to maintain energy balance. We previously described C1q/TNF-related protein 12 (CTRP12) as a novel metabolic hormone. Gain-of-function and partial-deficiency mouse models have highlighted important roles for this fat-derived adipokine in modulating systemic metabolism. Whether CTRP12 is essential and required for metabolic homeostasis is unknown. We show here that homozygous deletion of Ctrp12 gene results in sexually dimorphic phenotypes. Under basal conditions, complete loss of CTRP12 had little impact on male mice, whereas it decreased body weight (driven by reduced lean mass and liver weight) and improved insulin sensitivity in female mice. When challenged with a high-fat diet, Ctrp12 knockout (KO) male mice had decreased energy expenditure, increased weight gain and adiposity, elevated serum TNFα level, and reduced insulin sensitivity. In contrast, female KO mice had reduced weight gain and liver weight. The expression of lipid synthesis and catabolism genes, as well as profibrotic, endoplasmic reticulum stress, and oxidative stress genes were largely unaffected in the adipose tissue of Ctrp12 KO male mice. Despite greater adiposity and insulin resistance, Ctrp12 KO male mice fed an obesogenic diet had lower circulating triglyceride and free fatty acid levels. In contrast, lipid profiles of the leaner female KO mice were not different from those of WT controls. These data suggest that CTRP12 contributes to whole body energy metabolism in genotype-, diet-, and sex-dependent manners, underscoring complex gene-environment interactions influencing metabolic outcomes.
Collapse
Affiliation(s)
- Stefanie Y Tan
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xi Cao
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
47
|
Abstract
Purpose of Review In recent years, a family of adiponectin paralogs designated as C1q/TNF-related protein (CTRP) has attracted increasing attention. They are inflammatory adipocytokines mostly secreted from epicardial adipose tissue, which modulate the development and prognosis of coronary artery disease (CAD). This review summarizes the pathophysiological roles of individual members of the CTRP superfamily in the development of CAD. Recent Findings Recent studies have revealed how members of the CTRP family, CTRP1, CTRP3, CTRP5, CTRP9, CTRP12, and CTRP13, can influence both development and progression of CAD by modulating metabolic pathways, influencing immuno-inflammatory response, and regulating cardiovascular functions. Summary Research to date has not been sufficient to answer the specific mechanism of the CTRP family in the occurrence and development of CAD. This review explores the evidence of CTRP superfamily regulating different pathophysiology stages of CAD through the immuno-inflammation, glucose and lipid metabolism, and vascular endothelial function.
Collapse
Affiliation(s)
- Yueqiao Si
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Wenjun Fan
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China.
| |
Collapse
|
48
|
Zuo A, Li J, Zhao X, Li T, Lei S, Chen J, Xu D, Song C, Li N, Ruan S, Lyu L, Guo Y. Globular CTRP9 protects cardiomyocytes from palmitic acid-induced oxidative stress by enhancing autophagic flux. Chem Biol Interact 2020; 329:109094. [PMID: 32278738 DOI: 10.1016/j.cbi.2020.109094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/17/2020] [Accepted: 04/05/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Oxidative stress in cardiac myocytes is an important pathogenesis of cardiac lipotoxicity. Autophagy is a cellular self-digestion process that can selectively remove damaged organelles under oxidative stress, and thus presents a potential therapeutic target against cardiac lipotoxicity. Globular CTRP9 (gCTRP9) is a newly identified adiponectin paralog with established metabolic regulatory properties. The aim of this work is to investigate whether autophagy participates the protection effects of gCTRP9 in neonatal rat cardiac myocytes (NRCMs) under oxidative stress and the underlying mechanism. RESULTS NRCMs were treated with PA of various concentrations for indicated time period. Our results showed that PA enhanced intracellular ROS accumulation, decreased mitochondrial membrane potential (Δψm) and increased activation of caspases 3. These changes suggested lipotoxicity due to excessive PA. In addition, PA was observed to impair autophagic flux in NRCMs and impaired autophagosome clearance induced by PA contributes to cardiomyocyte death. Besides, we found that gCTRP9 increased the ratio of LC3II/I and the expression of ATG5 which was vital to the formation of autophagosomes and decreased the level of P62, suggesting enhanced autophagic flux in the absence or presence of PA. The result was further confirmed by the methods of infection with LC3-mRFP-GFP lentivirus and blockage of autophagosome-lysosome fusion by BafA1. Moreover, gCTRP9 reestablished the loss of mitochondrial membrane potential, suppressed ROS generation, and reduced PA -induced myocyte death. However, the protective effect of gCTRP9 on the cardiac lipotoxicity was partly abolished by blockade of autophagy by autophagy-related 5 (ATG5) siRNA, indicating that the effect of gCTRP9 on cell survival is critically mediated through regulation of autophagy. CONCLUSION Autophagy induction by gCTRP9 could be utilized as a potential therapeutic strategy against oxidative stress-mediated damage in cardiomyocytes.
Collapse
Affiliation(s)
- Anju Zuo
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Jun Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Xiaoyu Zhao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Clinical Trial Research Center, Jinan Central Hospital Affiliated to Shandong University, Ji'nan, People's Republic of China
| | - Tingting Li
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Shengyun Lei
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Jiying Chen
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Dan Xu
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Chengxiang Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Na Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Shiyan Ruan
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Linmao Lyu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Emergency, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China
| | - Yuan Guo
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China; Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, People's Republic of China.
| |
Collapse
|
49
|
Rodriguez S, Little HC, Daneshpajouhnejad P, Shepard BD, Tan SY, Wolfe A, Cheema MU, Jandu S, Woodward OM, Talbot CC, Berkowitz DE, Rosenberg AZ, Pluznick JL, Wong GW. Late-onset renal hypertrophy and dysfunction in mice lacking CTRP1. FASEB J 2020; 34:2657-2676. [PMID: 31908037 PMCID: PMC7739198 DOI: 10.1096/fj.201900558rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022]
Abstract
Local and systemic factors that influence renal structure and function in aging are not well understood. The secretory protein C1q/TNF-related protein 1 (CTRP1) regulates systemic metabolism and cardiovascular function. We provide evidence here that CTRP1 also modulates renal physiology in an age- and sex-dependent manner. In mice lacking CTRP1, we observed significantly increased kidney weight and glomerular hypertrophy in aged male but not female or young mice. Although glomerular filtration rate, plasma renin and aldosterone levels, and renal response to water restriction did not differ between genotypes, CTRP1-deficient male mice had elevated blood pressure. Echocardiogram and pulse wave velocity measurements indicated normal heart function and vascular stiffness in CTRP1-deficient animals, and increased blood pressure was not due to greater salt retention. Paradoxically, CTRP1-deficient mice had elevated urinary sodium and potassium excretion, partially resulting from reduced expression of genes involved in renal sodium and potassium reabsorption. Despite renal hypertrophy, markers of inflammation, fibrosis, and oxidative stress were reduced in CTRP1-deficient mice. RNA sequencing revealed alterations and enrichments of genes in metabolic processes in CTRP1-deficient animals. These results highlight novel contributions of CTRP1 to aging-associated changes in renal physiology.
Collapse
Affiliation(s)
- Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C. Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Blythe D. Shepard
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stefanie Y. Tan
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Muhammad Umar Cheema
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sandeep Jandu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Owen M. Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dan E. Berkowitz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jennifer L. Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G. William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
50
|
Zuo A, Zhao X, Li T, Li J, Lei S, Chen J, Xu D, Song C, Liu T, Li C, Guo Y. CTRP9 knockout exaggerates lipotoxicity in cardiac myocytes and high-fat diet-induced cardiac hypertrophy through inhibiting the LKB1/AMPK pathway. J Cell Mol Med 2020; 24:2635-2647. [PMID: 31930700 PMCID: PMC7028852 DOI: 10.1111/jcmm.14982] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/26/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022] Open
Abstract
CTRP9 has been reported to regulate lipid metabolism and exert cardioprotective effects, yet its role in high‐fat diet (HFD)‐induced cardiac lipotoxicity and the underlying mechanisms remain unclear. In the current study, we established HFD‐induced obesity model in wild‐type (WT) or CTRP9 knockout (CTRP9‐KO) mice and palmitate‐induced lipotoxicity model in neonatal rat cardiac myocytes (NRCMs) to investigate the effects of CTRP9 on cardiac lipotoxicity. Our results demonstrated that the HFD‐fed CTRP9‐KO mice accentuated cardiac hypertrophy, fibrosis, endoplasmic reticulum (ER) stress‐initiated apoptosis and oxidative stress compared with the HFD‐fed WT mice. In vitro, CTRP9 treatment markedly alleviated palmitate‐induced oxidative stress and ER stress‐induced apoptosis in NRCMs in a dose‐dependent manner. Phosphorylated AMPK at Thr172 was reduced, and phosphorylated mammalian target of rapamycin (mTOR) was strengthened in the heart of the HFD‐fed CTRP9‐KO mice compared with the HFD‐fed control mice. In vitro, AMPK inhibitor compound C significantly abolished the effects of CTRP9 on the inhibition of the apoptotic pathway in palmitate‐treated NRCMs. In a further mechanistic study, CTRP9 enhanced expression of phosphorylated LKB1 at Ser428 and promoted LKB1 cytoplasmic localization. Besides, silencing of LKB1 gene by lentivirus significantly prohibited activation of AMPK by CTRP9 and partially eliminated the protective effect of CTRP9 on the cardiac lipotoxicity. These results indicate that CTRP9 exerted anti‐myocardial lipotoxicity properties and inhibited cardiac hypertrophy probably through the LKB1/AMPK signalling pathway.
Collapse
Affiliation(s)
- Anju Zuo
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China
| | - Xiaoyu Zhao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Clinical Trial Research Center, Jinan Central Hospital Affiliated to Shandong University, Ji'nan, China
| | - Tingting Li
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Jun Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Shengyun Lei
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China
| | - Jiying Chen
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Dan Xu
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Chengxiang Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Tianjiao Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China
| | - Cuigang Li
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China
| | - Yuan Guo
- Department of General Medicine, Qilu Hospital of Shandong University, Ji'nan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Ji'nan, China
| |
Collapse
|