1
|
Li Q, Zhang H, Xiao N, Liang G, Lin Y, Yang X, Yang J, Qian Z, Fu Y, Zhang C, Liu A. Aging and Lifestyle Modifications for Preventing Aging-Related Diseases. FASEB J 2025; 39:e70575. [PMID: 40293686 DOI: 10.1096/fj.202402797rr] [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: 11/14/2024] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
The pathogenesis of various chronic diseases is closely associated with aging. Aging of the cardiovascular system promotes the development of severe cardiovascular diseases with high mortality, including atherosclerosis, coronary heart disease, and myocardial infarction. Similarly, aging of the nervous system promotes the development of neurodegenerative diseases, such as Alzheimer's disease, which seriously impairs cognitive function. Aging of the musculoskeletal system is characterized by decreased function and mobility. The molecular basis of organ aging is cellular senescence, which involves multiple cellular and molecular mechanisms, such as impaired autophagy, metabolic imbalance, oxidative stress, and persistent inflammation. Given the ongoing demographic shift toward an aging society, strategies to delay or reduce the effects of aging have gained significance. Lifestyle modifications, such as exercise and calorie restriction, are now recognized for their anti-aging effects, their capacity to reduce modification, their potential to prolong lifespan, and their capacity to lower the risk of cardiovascular disease. This review elucidates the molecular mechanisms and application significance of various anti-aging approaches at the molecular level, based on research progress in aging. It aims to provide a reference for the prevention and treatment of age-related diseases in progressively aging societies.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Chen Y, Xiao J, Zhu X, Fan X, Peng M, Mu Y, Wang C, Xia L, Zhou M. Exploiting conjugated linoleic acid for health: a recent update. Food Funct 2025; 16:147-167. [PMID: 39639784 DOI: 10.1039/d4fo04911j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Conjugated linoleic acid (CLA) is widely used as a dietary supplement due to its reported benefits in enhancing immunity, regulating inflammation, treating obesity, and preventing cancer. However, there is a lack of comprehensive studies on its mechanisms and dose-effect relationships. Moreover, there are insufficient in-depth studies on CLA's new functions, safety, side effects, and clinical utility. This review systematically examines the structure and sources of CLA, summarizes its role in improving human health, and critically reviews the potential mechanisms behind these benefits. It also analyzes the side effects of CLA and addresses issues related to dosing and oxidative decomposition in CLA research. Additionally, the potential of using CLA-producing probiotics to manage diseases is explored. This review can guide and promote further research on CLA's functions and support the development of CLA dietary supplements. It will accelerate the development of CLA nutritional and medical foods, contribute to the improvement of human health, and have important social meaning and economic value.
Collapse
Affiliation(s)
- Yang Chen
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Junfeng Xiao
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Xiaoqing Zhu
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Xin Fan
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Mingye Peng
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Yang Mu
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Chao Wang
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Lusha Xia
- Department of gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430068, China
| | - Mengzhou Zhou
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| |
Collapse
|
3
|
Sun S, Guo H, Chen G, Zhang H, Zhang Z, Wang X, Li D, Li X, Zhao G, Lin F. Peroxisome proliferator‑activated receptor γ coactivator‑1α in heart disease (Review). Mol Med Rep 2025; 31:17. [PMID: 39513608 PMCID: PMC11551696 DOI: 10.3892/mmr.2024.13382] [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: 07/29/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Heart disease (HD) is a general term for various diseases affecting the heart. An increasing body of evidence suggests that the pathogenesis of HD is closely related to mitochondrial dysfunction. Peroxisome proliferator‑activated receptor γ coactivator‑1α (PGC‑1α) is a transcriptional coactivator that plays an important role in mitochondrial function by regulating mitochondrial biogenesis, energy metabolism and oxidative stress. The present review shows that PGC‑1α expression and activity in the heart are controlled by multiple signaling pathways, including adenosine monophosphate‑activated protein kinase, sirtuin 1/3 and nuclear factor κB. These can mediate the activation or inhibition of transcription and post‑translational modifications (such as phosphorylation and acetylation) of PGC‑1α. Furthermore, it highlighted the recent progress of PGC‑1α in HD, including heart failure, coronary heart disease, diabetic cardiomyopathy, drug‑induced cardiotoxicity and arrhythmia. Understanding the mechanisms underlying PGC‑1α in response to pathological stimulation may prove to be beneficial in developing new ideas and strategies for preventing and treating HDs. Meanwhile, the present review explored why the opposite results occurred when PGC‑1α was used as a target therapy.
Collapse
Affiliation(s)
- Siyu Sun
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Huige Guo
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang, Henan 453000, P.R. China
| | - Guohui Chen
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Hui Zhang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Zhanrui Zhang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Xiulong Wang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Dongxu Li
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Xuefang Li
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Guoan Zhao
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Fei Lin
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| |
Collapse
|
4
|
Saki N, Haybar H, Maniati M, Davari N, Javan M, Moghimian-Boroujeni B. Modification macrophage to foam cells in atherosclerosis disease: some factors stimulate or inhibit this process. J Diabetes Metab Disord 2024; 23:1687-1697. [PMID: 39610485 PMCID: PMC11599683 DOI: 10.1007/s40200-024-01482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/16/2024] [Indexed: 11/30/2024]
Abstract
Background Atherosclerosis is an arterial blood vessel disease that begins and progresses by turning macrophages into foam cells. Uptake of oxidized low-density lipoprotein (ox-LDL), cholesterol esterification and cholesterol efflux are the most important factors in the formation of foam cells and play an important role in atherosclerosis. Methods The present study is based on the data obtained from the PubMed database (1961-2024) using the MeSH search terms "Atherosclerosis", "Macrophages" and "Foam cells". Reviews for writing the main text and non-English-language articles were excluded. Result The interaction between ox-LDL and macrophages plays an important role in plaque initiation and promotion processes. Macrophages abnormally digest ox-LDL, resulting in the accumulation of lipids and formation of foam cells. This is an important step in the development of atherosclerosis. Also, several other factors such as inflammatory factors, growth factors, hormones, etc. can play an important role in the development of atherosclerotic lesions or counteract it by affecting the formation of foam cells. Conclusion Several factors can affect the progression of atherosclerosis by affecting macrophage activity or its conversion to foam cells. Also, some of these factors play a protective role against the development and atherosclerosis progression. In this paper, we reviewed some of these factors and their effect on atherosclerosis.
Collapse
Affiliation(s)
- Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Habib Haybar
- Cardiology Department, Medical College, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmood Maniati
- School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nader Davari
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadreza Javan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Bahareh Moghimian-Boroujeni
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Lin S, Hou L, Wang Y, Lin H, Deng J, Li S, Long H, Zhao G. Antagonism of let-7c reduces atherosclerosis and macrophage lipid accumulation by promoting PGC-1α/LXRα/ABCA1/G1 pathway. Gene 2024; 909:148302. [PMID: 38401833 DOI: 10.1016/j.gene.2024.148302] [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: 11/20/2023] [Revised: 02/05/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
Changes in circulating let-7c were significantly associated with the alter in lipid profile, but its role in intracellular lipid metabolism remains unknown. This work was conducted to explore the effects of let-7c on the lipid accumulation in macrophages and uncover the underlying mechanism. Our results showed that let-7c inhibition relieved atherosclerosis progression in apoE-/- mice. In ox-LDL-treatment macrophages, let-7c knockdown suppressed lipid accumulation but does no affect cholesterol intake. Consistent with this, overexpression of let-7c promoted lipid accumulation by reducing the expression of LXRα and ABCA1/G1. Mechanistically, let-7c targeted PGC-1α to repress the expression of LXRα and ABCA1/G1, thereby regulating cholesterol homeostasis in macrophages. Taken together, these findings suggest that antagonism of let-7c reduces atherosclerosis and macrophage lipid accumulation through the PGC-1α/LXRα/ABCA1/G1 axis.
Collapse
Affiliation(s)
- Shuyun Lin
- Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511500, China
| | - Lianjie Hou
- Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511500, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511500, China
| | - Huiling Lin
- Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Jiefeng Deng
- College of Pharmacy, Dali University, Dali 671000, China
| | - Shuang Li
- College of Pharmacy, Dali University, Dali 671000, China
| | - Haijiao Long
- Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511500, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511500, China.
| |
Collapse
|
6
|
Feng X, Tong L, Ma L, Mu T, Yu B, Ma R, Li J, Wang C, Zhang J, Gu Y. Mining key circRNA-associated-ceRNA networks for milk fat metabolism in cows with varying milk fat percentages. BMC Genomics 2024; 25:323. [PMID: 38561663 PMCID: PMC10983688 DOI: 10.1186/s12864-024-10252-y] [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: 08/31/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Cow milk fat is an essential indicator for evaluating and measuring milk quality and cow performance. Growing research has identified the molecular functions of circular RNAs (circRNAs) necessary for mammary gland development and lactation in mammals. METHOD The present study analyzed circRNA expression profiling data in mammary epithelial cells (MECs) from cows with highly variable milk fat percentage (MFP) using differential expression analysis and weighted gene co-expression network analysis (WGCNA). RESULTS A total of 309 differentially expressed circRNAs (DE-circRNAs) were identified in the high and low MFP groups. WGCNA analysis revealed that the pink module was significantly associated with MFP (r = - 0.85, P = 0.007). Parental genes of circRNAs in this module were enriched mainly in lipid metabolism-related signaling pathways, such as focal adhesion, ECM-receptor interaction, adherens junction and AMPK. Finally, six DE-circRNAs were screened from the pink module: circ_0010571, circ_0007797, circ_0002746, circ_0003052, circ_0004319, and circ_0012840. Among them, circ_0002746, circ_0003052, circ_0004319, and circ_0012840 had circular structures and were highly expressed in mammary tissues. Subcellular localization revealed that these four DE-circRNAs may play a regulatory role in the mammary glands of dairy cows, mainly as competitive endogenous RNAs (ceRNAs). Seven hub target genes (GNB1, GNG2, PLCB1, PLCG1, ATP6V0C, NDUFS4, and PIGH) were obtained by constructing the regulatory network of their ceRNAs and then analyzed by CytoHubba and MCODE plugins in Cytoscape. Functional enrichment analysis revealed that these genes are crucial and most probable ceRNA regulators in milk fat metabolism. CONCLUSIONS Our study identified several vital circRNAs and ceRNAs affecting milk fat synthesis, providing new research ideas and a theoretical basis for cow lactation, milk quality, and breed improvement.
Collapse
Affiliation(s)
- Xiaofang Feng
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Lijia Tong
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Lina Ma
- NingXia Academy of Agriculture and Forestry Sciences, 750002, Yinchuan, China
| | - Tong Mu
- School of Life Science, Yan'an University, 716000, Yanan, China
| | - Baojun Yu
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Ruoshuang Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Jiwei Li
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Chuanchuan Wang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Juan Zhang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China.
| | - Yaling Gu
- Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, 750021, Yinchuan, China
| |
Collapse
|
7
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 76] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
8
|
Castoldi A, Sanin DE, van Teijlingen Bakker N, Aguiar CF, de Brito Monteiro L, Rana N, Grzes KM, Kabat AM, Curtis J, Cameron AM, Caputa G, Antônio de Souza T, Souto FO, Buescher JM, Edwards-Hicks J, Pearce EL, Pearce EJ, Saraiva Camara NO. Metabolic and functional remodeling of colonic macrophages in response to high-fat diet-induced obesity. iScience 2023; 26:107719. [PMID: 37674984 PMCID: PMC10477064 DOI: 10.1016/j.isci.2023.107719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/17/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
Little is known about the effects of high-fat diet (HFD)-induced obesity on resident colonic lamina propria (LP) macrophages (LPMs) function and metabolism. Here, we report that obesity and diabetes resulted in increased macrophage infiltration in the colon. These macrophages exhibited the residency phenotype CX3CR1hiMHCIIhi and were CD4-TIM4-. During HFD, resident colonic LPM exhibited a lipid metabolism gene expression signature that overlapped that used to define lipid-associated macrophages (LAMs). Via single-cell RNA sequencing, we identified a sub-cluster of macrophages, increased in HFD, that were responsible for the LAM signature. Compared to other macrophages in the colon, these cells were characterized by elevated glycolysis, phagocytosis, and efferocytosis signatures. CX3CR1hiMHCIIhi colonic resident LPMs had fewer lipid droplets (LDs) and decreased triacylglycerol (TG) content compared to equivalent cells in lean mice and exhibited increased phagocytic capacity, suggesting that HFD induces adaptive responses in LPMs to limit bacterial translocation.
Collapse
Affiliation(s)
- Angela Castoldi
- Department of Immunology, University of Sao Paulo, Sao Paulo, Brazil
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Institute Keizo Asami, Federal University of Pernambuco, Pernambuco, Brazil
| | - David E. Sanin
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Nikki van Teijlingen Bakker
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | | | - Lauar de Brito Monteiro
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Nisha Rana
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Katarzyna M. Grzes
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Agnieszka M. Kabat
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jonathan Curtis
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alanna M. Cameron
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - George Caputa
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | | | - Fabrício O. Souto
- Institute Keizo Asami, Federal University of Pernambuco, Pernambuco, Brazil
| | - Joerg M. Buescher
- Metabolomics Facility, Max Planck Institute of Epigenetics and Immunobiology, Freiburg im Breisgau, Germany
| | - Joy Edwards-Hicks
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Erika L. Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Edward J. Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | |
Collapse
|
9
|
Gindri dos Santos B, Goedeke L. Macrophage immunometabolism in diabetes-associated atherosclerosis. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00032. [PMID: 37849988 PMCID: PMC10578522 DOI: 10.1097/in9.0000000000000032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023]
Abstract
Macrophages play fundamental roles in atherosclerotic plaque formation, growth, and regression. These cells are extremely plastic and perform different immune functions depending on the stimuli they receive. Initial in vitro studies have identified specific metabolic pathways that are crucial for the proper function of pro-inflammatory and pro-resolving macrophages. However, the plaque microenvironment, especially in the context of insulin resistance and type 2 diabetes, constantly challenges macrophages with several simultaneous inflammatory and metabolic stimuli, which may explain why atherosclerosis is accelerated in diabetic patients. In this mini review, we discuss how macrophage mitochondrial function and metabolism of carbohydrates, lipids, and amino acids may be affected by this complex plaque microenvironment and how risk factors associated with type 2 diabetes alter the metabolic rewiring of macrophages and disease progression. We also briefly discuss current challenges in assessing macrophage metabolism and identify future tools and possible strategies to alter macrophage metabolism to improve treatment options for diabetes-associated atherosclerosis.
Collapse
Affiliation(s)
- Bernardo Gindri dos Santos
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leigh Goedeke
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine (Endocrinology), The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
10
|
Lee M, Lee SY, Bae YS. Functional roles of sphingolipids in immunity and their implication in disease. Exp Mol Med 2023; 55:1110-1130. [PMID: 37258585 PMCID: PMC10318102 DOI: 10.1038/s12276-023-01018-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 06/02/2023] Open
Abstract
Sphingolipids, which are components of cellular membranes and organ tissues, can be synthesized or degraded to modulate cellular responses according to environmental cues, and the balance among the different sphingolipids is important for directing immune responses, regardless of whether they originate, as intra- or extracellular immune events. Recent progress in multiomics-based analyses and methodological approaches has revealed that human health and diseases are closely related to the homeostasis of sphingolipid metabolism, and disease-specific alterations in sphingolipids and related enzymes can be prognostic markers of human disease progression. Accumulating human clinical data from genome-wide association studies and preclinical data from disease models provide support for the notion that sphingolipids are the missing pieces that supplement our understanding of immune responses and diseases in which the functions of the involved proteins and nucleotides have been established. In this review, we analyze sphingolipid-related enzymes and reported human diseases to understand the important roles of sphingolipid metabolism. We discuss the defects and alterations in sphingolipid metabolism in human disease, along with functional roles in immune cells. We also introduce several methodological approaches and provide summaries of research on sphingolipid modulators in this review that should be helpful in studying the roles of sphingolipids in preclinical studies for the investigation of experimental and molecular medicines.
Collapse
Affiliation(s)
- Mingyu Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Suh Yeon Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yoe-Sik Bae
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
11
|
Barbero NM, Oller J, Sanz AB, Ramos AM, Ortiz A, Ruiz-Ortega M, Rayego-Mateos S. Mitochondrial Dysfunction in the Cardio-Renal Axis. Int J Mol Sci 2023; 24:ijms24098209. [PMID: 37175915 PMCID: PMC10179675 DOI: 10.3390/ijms24098209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Cardiovascular disease (CVD) frequently complicates chronic kidney disease (CKD). The risk of all-cause mortality increases from 20% to 500% in patients who suffer both conditions; this is referred to as the so-called cardio-renal syndrome (CRS). Preclinical studies have described the key role of mitochondrial dysfunction in cardiovascular and renal diseases, suggesting that maintaining mitochondrial homeostasis is a promising therapeutic strategy for CRS. In this review, we explore the malfunction of mitochondrial homeostasis (mitochondrial biogenesis, dynamics, oxidative stress, and mitophagy) and how it contributes to the development and progression of the main vascular pathologies that could be affected by kidney injury and vice versa, and how this knowledge may guide the development of novel therapeutic strategies in CRS.
Collapse
Affiliation(s)
- Nerea Mendez Barbero
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Jorge Oller
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Ana B Sanz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Adrian M Ramos
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Marta Ruiz-Ortega
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| | - Sandra Rayego-Mateos
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| |
Collapse
|
12
|
Qin HL, Bao JH, Tang JJ, Xu DY, Shen L. Arterial remodeling: the role of mitochondrial metabolism in vascular smooth muscle cells. Am J Physiol Cell Physiol 2023; 324:C183-C192. [PMID: 36468843 DOI: 10.1152/ajpcell.00074.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Arterial remodeling is a common pathological basis of cardiovascular diseases such as atherosclerosis, vascular restenosis, hypertension, pulmonary hypertension, aortic dissection, and aneurysm. Vascular smooth muscle cells (VSMCs) are not only the main cellular components in the middle layer of the arterial wall but also the main cells involved in arterial remodeling. Dedifferentiated VSMCs lose their contractile properties and are converted to a synthetic, secretory, proliferative, and migratory phenotype, playing key roles in the pathogenesis of arterial remodeling. As mitochondria are the main site of biological oxidation and energy transformation in eukaryotic cells, mitochondrial numbers and function are very important in maintaining the metabolic processes in VSMCs. Mitochondrial dysfunction and oxidative stress are novel triggers of the phenotypic transformation of VSMCs, leading to the onset and development of arterial remodeling. Therefore, pharmacological measures that alleviate mitochondrial dysfunction reverse arterial remodeling by ameliorating VSMCs metabolic dysfunction and phenotypic transformation, providing new options for the treatment of cardiovascular diseases related to arterial remodeling. This review summarizes the relationship between mitochondrial dysfunction and cardiovascular diseases associated with arterial remodeling and then discusses the potential mechanism by which mitochondrial dysfunction participates in pathological arterial remodeling. Furthermore, maintaining or improving mitochondrial function may be a new intervention strategy to prevent the progression of arterial remodeling.
Collapse
Affiliation(s)
- Hua-Li Qin
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing-Hui Bao
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jian-Jun Tang
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Dan-Yan Xu
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Shen
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Sottero B, Testa G, Gamba P, Staurenghi E, Giannelli S, Leonarduzzi G. Macrophage polarization by potential nutraceutical compounds: A strategic approach to counteract inflammation in atherosclerosis. Free Radic Biol Med 2022; 181:251-269. [PMID: 35158030 DOI: 10.1016/j.freeradbiomed.2022.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022]
Abstract
Chronic inflammation represents a main event in the onset and progression of atherosclerosis and is closely associated with oxidative stress in a sort of vicious circle that amplifies and sustains all stages of the disease. Key players of atherosclerosis are monocytes/macrophages. According to their pro- or anti-inflammatory phenotype and biological functions, lesional macrophages can release various mediators and enzymes, which in turn contribute to plaque progression and destabilization or, alternatively, lead to its resolution. Among the factors connected to atherosclerotic disease, lipid species carried by low density lipoproteins and pro-oxidant stimuli strongly promote inflammatory events in the vasculature, also by modulating the macrophage phenotyping. Therapies specifically aimed to balance macrophage inflammatory state are increasingly considered as powerful tools to counteract plaque formation and destabilization. In this connection, several molecules of natural origin have been recognized to be active mediators of diverse metabolic and signaling pathways regulating lipid homeostasis, redox state, and inflammation; they are, thus, considered as promising candidates to modulate macrophage responsiveness to pro-atherogenic stimuli. The current knowledge of the capability of nutraceuticals to target macrophage polarization and to counteract atherosclerotic lesion progression, based mainly on in vitro investigation, is summarized in the present review.
Collapse
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy.
| |
Collapse
|
14
|
Gao Z, Xu X, Li Y, Sun K, Yang M, Zhang Q, Wang S, Lin Y, Lou L, Wu A, Liu W, Nie B. Mechanistic Insight into PPARγ and Tregs in Atherosclerotic Immune Inflammation. Front Pharmacol 2021; 12:750078. [PMID: 34658891 PMCID: PMC8511522 DOI: 10.3389/fphar.2021.750078] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/17/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis (AS) is the main pathological cause of acute cardiovascular and cerebrovascular diseases, such as acute myocardial infarction and cerebral apoplexy. As an immune-mediated inflammatory disease, the pathogenesis of AS involves endothelial cell dysfunction, lipid accumulation, foam cell formation, vascular smooth muscle cell (VSMC) migration, and inflammatory factor infiltration. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) plays an important role in lipid metabolism, inflammation, and apoptosis by antagonizing the Wnt/β-catenin pathway and regulating cholesterol efflux and inflammatory factors. Importantly, PPARγ-dependant fatty acid uptake is critical for metabolic programming. Activated PPARγ can exert an anti-atherosclerotic effect by inhibiting the expression of various inflammatory factors, improving endothelial cell function, and restraining the proliferation and migration of VSMCs. Regulatory T cells (Tregs) are the only subset of T lymphocytes that have a completely negative regulatory effect on the autoimmune response. They play a critical role in suppressing excessive immune responses and inflammatory reactions and widely affect AS-associated foam cell formation, plaque rupture, and other processes. Recent studies have shown that PPARγ activation promotes the recruitment of Tregs to reduce inflammation, thereby exerting its anti-atherosclerotic effect. In this review, we provide an overview of the anti-AS roles of PPARγ and Tregs by discussing their pathological mechanisms from the perspective of AS and immune-mediated inflammation, with a focus on basic research and clinical trials of their efficacies alone or in combination in inhibiting atherosclerotic inflammation. Additionally, we explore new ideas for AS treatment and plaque stabilization and establish a foundation for the development of natural PPARγ agonists with Treg recruitment capability.
Collapse
Affiliation(s)
- Zhao Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China.,Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Institute of Nephrology, Guangdong Medical University, Zhanjiang, China
| | - Xinrui Xu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Yang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kehan Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Manfang Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyue Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Shuqi Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiyi Lin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Lixia Lou
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Aiming Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China.,Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Institute of Nephrology, Guangdong Medical University, Zhanjiang, China
| | - Bo Nie
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Morroniside Promotes PGC-1 α-Mediated Cholesterol Efflux in Sodium Palmitate or High Glucose-Induced Mouse Renal Tubular Epithelial Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9942152. [PMID: 34485530 PMCID: PMC8410408 DOI: 10.1155/2021/9942152] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 08/05/2021] [Indexed: 12/26/2022]
Abstract
Lipid deposition is an etiology of renal damage caused by lipid metabolism disorder in diabetic nephropathy (DN). Thus, reducing lipid deposition is a feasible strategy for the treatment of DN. Morroniside (MOR), an iridoid glycoside isolated from the Chinese herb Cornus officinalis Sieb. et Zucc., is considered to be an effective drug in inhibiting oxidative stress, reducing inflammatory response, and countering apoptosis. To explore the protective mechanism of MOR in attenuating renal lipotoxicity in DN, we investigated the effect of MOR on an in vitro model of lipid metabolism disorder of DN established by stimulating mouse renal tubular epithelial cells (mRTECs) with sodium palmitate (PA) or high glucose (HG). Oil Red O and filipin cholesterol staining assays were used to determine intracellular lipid accumulation status. Results revealed that PA or HG stimulation inhibited the expressions of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), liver X receptors (LXR), ATP-binding cassette subfamily A member 1 (ABCA1), ABCG1, and apolipoprotein E (ApoE) in mRTECs as evidenced by western blot and quantitative real-time PCR, resulting in increased intracellular lipid deposition. Interestingly, MOR upregulated expressions of PGC-1α, LXR, ABCA1, ABCG1, and ApoE, thus reducing cholesterol accumulation in mRTECs, suggesting that MOR might promote cholesterol efflux from mRTECs via the PGC-1α/LXR pathway. Of note, silencing PGC-1α reversed the promotive effect of MOR on PA- or HG-induced cellular cholesterol accumulation. In conclusion, our results suggest that MOR has a protective effect on mRTECs under high lipid or high glucose conditions, which may be related to the promotion of intracellular cholesterol efflux mediated by PGC-1α.
Collapse
|
16
|
Rojas MM, Villalpando DM, Alexander-Aguilera A, Ferrer M, García HS. Effect of CLA supplementation on factors related to vascular dysfunction in arteries of orchidectomized rats. Prostaglandins Other Lipid Mediat 2021; 157:106586. [PMID: 34438054 DOI: 10.1016/j.prostaglandins.2021.106586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 01/22/2023]
Abstract
The vascular endothelium is a monolayer of flat epithelial cells located between the circulating blood and the underlying connective tissue. It conveys key functions that when impaired, lead to endothelial dysfunction. This condition is responsible for the pathogenesis of vascular diseases. The cardioprotective effect of sex hormones is widely known; hence, a murine orchidectomized model has been employed to study the effects caused by their deficiency. In the search for approaches to maintain vascular health, the effect of dietary fatty acids as CLA on cardiovascular diseases has been studied. Some proven beneficial properties of CLA are antioxidant, antiatherogenic and anti-inflammatory. Our objective was to evaluate the effect of a diet supplemented with 1.8 % (w/w) of CLA, administered during eight weeks, on the amount of cholesterol oxidation products (COPs) produced by orchidectomy and on factors related to vascular dysfunction in the aorta and the mesenteric arteries. The diet with CLA prevented the increase in prostanoids formation and maintained the normal physiological conditions of NO and antioxidant activity. In addition, it prevented the increase in cholesterol and COPs at the vascular wall. CLA-supplemented diet prevented the orchidectomy-induced alterations on prostanoids, NO and COPs and also improved the antioxidant activity. These findings could contribute to understand the mechanisms of actions of CLA involved in the prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Mibsam M Rojas
- Food Research and Development Unit, National Technology of Mexico/Technological Institute of Veracruz, Ver., Mexico
| | - Diva M Villalpando
- Physiology Department, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | | | - Mercedes Ferrer
- Physiology Department, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain.
| | - Hugo S García
- Food Research and Development Unit, National Technology of Mexico/Technological Institute of Veracruz, Ver., Mexico.
| |
Collapse
|
17
|
Wei Z, Chong H, Jiang Q, Tang Y, Xu J, Wang H, Shi Y, Cui L, Li J, Zhang Y, Xue Y, Li J, Liu G, Chen X, Wang D, Zhang CY, Jiang X. Smooth Muscle Overexpression of PGC1α Attenuates Atherosclerosis in Rabbits. Circ Res 2021; 129:e72-e86. [PMID: 34162227 DOI: 10.1161/circresaha.120.317705] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zhe Wei
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Hoshun Chong
- Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, China (H.C., Y.X.)
| | - Qixia Jiang
- Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China (Q.J., Jutang Li)
| | - Yuhang Tang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Jinhong Xu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Haoquan Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Yanteng Shi
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Le Cui
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Jing Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Yujing Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Yunxing Xue
- Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, China (H.C., Y.X.)
| | - Jutang Li
- Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China (Q.J., Jutang Li)
| | - George Liu
- Institute of Cardiovascular Science, Peking University, Beijing, China (G.L.)
| | - Xi Chen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Dongjin Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Xiaohong Jiang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| |
Collapse
|
18
|
Yuan Y, Yuan L, Li L, Liu F, Liu J, Chen Y, Cheng J, Lu Y. Mitochondrial transfer from mesenchymal stem cells to macrophages restricts inflammation and alleviates kidney injury in diabetic nephropathy mice via PGC-1α activation. STEM CELLS (DAYTON, OHIO) 2021; 39:913-928. [PMID: 33739541 DOI: 10.1002/stem.3375] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/23/2021] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs) have fueled ample translation for treatment of immune-mediated diseases. Our previous study had demonstrated that MSCs could elicit macrophages (Mφ) into anti-inflammatory phenotypes, and alleviate kidney injury in diabetic nephropathy (DN) mice via improving mitochondrial function of Mφ, yet the specific mechanism was unclear. Recent evidence indicated that MSCs communicated with their microenvironment through exchanges of mitochondria. By a coculture system consisting of MSCs and Mφ, we showed that MSCs-derived mitochondria (MSCs-Mito) were transferred into Mφ, and the mitochondrial functions were improved, which contributed to M2 polarization. Furthermore, we found that MSCs-Mito transfer activated peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α)-mediated mitochondrial biogenesis. In addition, PGC-1α interacted with TFEB in high glucose-induced Mφ, leading to the elevated lysosome-autophagy, which was essential to removal of damaged mitochondria. As a result, in Mφ, the mitochondrial bioenergy and capacity to combat inflammatory response were enhanced. Whereas, the immune-regulatory activity of MSCs-Mito was significantly blocked in PGC-1α knockdown Mφ. More importantly, MSCs-Mito transfer could be observed in DN mice, and the adoptive transfer of MSCs-Mito educated Mφ (MφMito ) inhibited the inflammatory response and alleviated kidney injury. However, the kidney-protective effects of MφMito were abolished when the MSCs-Mito was impaired with rotenone, and the similar results were also observed when MφMito were transfected with sipgc-1α before administration. Collectively, these findings suggested that MSCs elicited Mφ into anti-inflammatory phenotype and ameliorated kidney injury through mitochondrial transfer in DN mice, and the effects were relied on PGC-1α-mediated mitochondrial biogenesis and PGC-1α/TFEB-mediated lysosome-autophagy.
Collapse
Affiliation(s)
- Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Longhui Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Fei Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
19
|
Interface of Phospholipase Activity, Immune Cell Function, and Atherosclerosis. Biomolecules 2020; 10:biom10101449. [PMID: 33076403 PMCID: PMC7602611 DOI: 10.3390/biom10101449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 09/30/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
Phospholipases are a family of lipid-altering enzymes that can either reduce or increase bioactive lipid levels. Bioactive lipids elicit signaling responses, activate transcription factors, promote G-coupled-protein activity, and modulate membrane fluidity, which mediates cellular function. Phospholipases and the bioactive lipids they produce are important regulators of immune cell activity, dictating both pro-inflammatory and pro-resolving activity. During atherosclerosis, pro-inflammatory and pro-resolving activities govern atherosclerosis progression and regression, respectively. This review will look at the interface of phospholipase activity, immune cell function, and atherosclerosis.
Collapse
|
20
|
Gerbec ZJ, Hashemi E, Nanbakhsh A, Holzhauer S, Yang C, Mei A, Tsaih SW, Lemke A, Flister MJ, Riese MJ, Thakar MS, Malarkannan S. Conditional Deletion of PGC-1α Results in Energetic and Functional Defects in NK Cells. iScience 2020; 23:101454. [PMID: 32858341 PMCID: PMC7474003 DOI: 10.1016/j.isci.2020.101454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/30/2019] [Accepted: 08/10/2020] [Indexed: 01/07/2023] Open
Abstract
During an immune response, natural killer (NK) cells activate specific metabolic pathways to meet the increased energetic and biosynthetic demands associated with effector functions. Here, we found in vivo activation of NK cells during Listeria monocytogenes infection-augmented transcription of genes encoding mitochondria-associated proteins in a manner dependent on the transcriptional coactivator PGC-1α. Using an Ncr1Cre-based conditional knockout mouse, we found that PGC-1α was crucial for optimal NK cell effector functions and bioenergetics, as the deletion of PGC-1α was associated with decreased cytotoxic potential and cytokine production along with altered ADP/ATP ratios. Lack of PGC-1α also significantly impaired the ability of NK cells to control B16F10 tumor growth in vivo, and subsequent gene expression analysis showed that PGC-1α mediates transcription required to maintain mitochondrial activity within the tumor microenvironment. Together, these data suggest that PGC-1α-dependent transcription of specific target genes is required for optimal NK cell function during the response to infection or tumor growth.
Collapse
Affiliation(s)
- Zachary J. Gerbec
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arash Nanbakhsh
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Sandra Holzhauer
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Chao Yang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ao Mei
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shirng-Wern Tsaih
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Angela Lemke
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael J. Flister
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monica S. Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
21
|
Mohammadi I, Mahdavi AH, Rabiee F, Nasr Esfahani MH, Ghaedi K. Positive effects of conjugated linoleic acid (CLA) on the PGC1-α expression under the inflammatory conditions induced by TNF-α in the C2C12 cell line. Gene 2020; 735:144394. [DOI: 10.1016/j.gene.2020.144394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/17/2022]
|
22
|
Mouton AJ, Li X, Hall ME, Hall JE. Obesity, Hypertension, and Cardiac Dysfunction: Novel Roles of Immunometabolism in Macrophage Activation and Inflammation. Circ Res 2020; 126:789-806. [PMID: 32163341 PMCID: PMC7255054 DOI: 10.1161/circresaha.119.312321] [Citation(s) in RCA: 363] [Impact Index Per Article: 72.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity and hypertension, which often coexist, are major risk factors for heart failure and are characterized by chronic, low-grade inflammation, which promotes adverse cardiac remodeling. While macrophages play a key role in cardiac remodeling, dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypes promotes excessive inflammation and cardiac injury. Metabolic shifting between glycolysis and mitochondrial oxidative phosphorylation has been implicated in macrophage polarization. M1 macrophages primarily rely on glycolysis, whereas M2 macrophages rely on the tricarboxylic acid cycle and oxidative phosphorylation; thus, factors that affect macrophage metabolism may disrupt M1/M2 homeostasis and exacerbate inflammation. The mechanisms by which obesity and hypertension may synergistically induce macrophage metabolic dysfunction, particularly during cardiac remodeling, are not fully understood. We propose that obesity and hypertension induce M1 macrophage polarization via mechanisms that directly target macrophage metabolism, including changes in circulating glucose and fatty acid substrates, lipotoxicity, and tissue hypoxia. We discuss canonical and novel proinflammatory roles of macrophages during obesity-hypertension-induced cardiac injury, including diastolic dysfunction and impaired calcium handling. Finally, we discuss the current status of potential therapies to target macrophage metabolism during heart failure, including antidiabetic therapies, anti-inflammatory therapies, and novel immunometabolic agents.
Collapse
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - Michael E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| | - John E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street; Jackson, MS, 39216-4505
| |
Collapse
|
23
|
Lee G, Uddin MJ, Kim Y, Ko M, Yu I, Ha H. PGC-1α, a potential therapeutic target against kidney aging. Aging Cell 2019; 18:e12994. [PMID: 31313501 PMCID: PMC6718532 DOI: 10.1111/acel.12994] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is defined as changes in an organism over time. The proportion of the aged population is markedly increasing worldwide. The kidney, as an essential organ with a high energy requirement, is one of the most susceptible organs to aging. It is involved in glucose metabolism via gluconeogenesis, glucose filtration and reabsorption, and glucose utilization. Proximal tubular epithelial cells (PTECs) depend on lipid metabolism to meet the high demand for ATP. Recent studies have shown that aging‐related kidney dysfunction is highly associated with metabolic changes in the kidney. Peroxisome proliferator‐activated receptor gamma coactivator‐1 alpha (PGC‐1α), a transcriptional coactivator, plays a major role in the regulation of mitochondrial biogenesis, peroxisomal biogenesis, and glucose and lipid metabolism. PGC‐1α is abundant in tissues, including kidney PTECs, which demand high energy. Many in vitro and in vivo studies have demonstrated that the activation of PGC‐1α by genetic or pharmacological intervention prevents telomere shortening and aging‐related changes in the skeletal muscle, heart, and brain. The activation of PGC‐1α can also prevent kidney dysfunction in various kidney diseases. Therefore, a better understanding of the effect of PGC‐1α activation in various organs on aging and kidney diseases may unveil a potential therapeutic strategy against kidney aging.
Collapse
Affiliation(s)
- Gayoung Lee
- Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Md Jamal Uddin
- Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Yoojeong Kim
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Minji Ko
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Inyoung Yu
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
- College of Pharmacy Ewha Womans University Seoul Korea
| |
Collapse
|
24
|
Chen YF, Dugas TR. Endothelial mitochondrial senescence accelerates cardiovascular disease in antiretroviral-receiving HIV patients. Toxicol Lett 2019; 317:13-23. [PMID: 31562912 DOI: 10.1016/j.toxlet.2019.09.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/21/2019] [Indexed: 02/06/2023]
Abstract
Combination antiretroviral therapy (cART) has been hugely successful in reducing the mortality associated with human immunodeficiency virus (HIV) infection, resulting in a growing population of people living with HIV (PLWH). Since PLWH now have a longer life expectancy, chronic comorbidities have become the focus of the clinical management of HIV. For example, cardiovascular complications are now one of the most prevalent causes of death in PLWH. Numerous epidemiological studies show that antiretroviral treatment increases cardiovascular disease (CVD) risk and early onset of CVD in PLWH. Nucleoside reverse transcriptase inhibitors (NRTIs) are the backbone of cART, and two NRTIs are typically used in combination with one drug from another drug class, e.g., a fusion inhibitor. NRTIs are known to induce mitochondrial dysfunction, contributing to toxicity in numerous tissues, such as myopathy, lipoatrophy, neuropathy, and nephropathy. In in vitro studies, short-term NRTI treatment induces an endothelial dysfunction with an increased reactive oxygen species (ROS) production; long-term NRTI treatment decreases cell replication capacity, while increasing mtROS production and senescent cell accumulation. These findings suggest that a mitochondrial oxidative stress is involved in the pathogenesis of NRTI-induced endothelial dysfunction and premature senescence. Mitochondrial dysfunction, defined by a compromised mitochondrial quality control via biogenesis and mitophagy, has a causal role in premature endothelial senescence and can potentially initiate early cardiovascular disease (CVD) development in PLWH. In this review, we explore the hypothesis and present literature supporting that long-term NRTI treatment induces vascular dysfunction by interfering with endothelial mitochondrial homeostasis and provoking mitochondrial genomic instability, resulting in premature endothelial senescence.
Collapse
Affiliation(s)
- Yi-Fan Chen
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, United States
| | - Tammy R Dugas
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, United States.
| |
Collapse
|
25
|
Bruen R, Curley S, Kajani S, Lynch G, O'Reilly ME, Dillon ET, Fitzsimons S, Mthunzi L, McGillicuddy FC, Belton O. Different monocyte phenotypes result in proresolving macrophages in conjugated linoleic acid-induced attenuated progression and regression of atherosclerosis. FASEB J 2019; 33:11006-11020. [PMID: 31284764 DOI: 10.1096/fj.201900922r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Monocytes/macrophages drive progression and regression of atherosclerosis. Conjugated linoleic acid (CLA), an anti-inflammatory lipid, mediates atheroprotective effects. We investigated how CLA alters monocyte/macrophage phenotype during attenuated progression and regression of atherosclerosis. Apolipoprotein E knockout (ApoE-/-) mice were fed a high-fat (60%) high-cholesterol (1%) diet (HFHCD) for 2 wk, followed by 6-wk 1% CLA 80:20 supplementation to investigate disease progression. Simultaneously, ApoE-/- mice were fed a 12-wk HFHCD with/without CLA for the final 4 wk to investigate regression. Aortic lesions were quantified by en face staining. Proteomic analysis, real-time quantitative PCR and flow cytometry were used to interrogate monocyte/macrophage phenotypes. CLA supplementation inhibited atherosclerosis progression coincident with decreased proinflammatory and increased anti-inflammatory macrophages. However, CLA-induced regression was associated with increased proinflammatory monocytes resulting in increased proresolving M2 bone marrow-derived macrophages, splenic macrophages, and dendritic cells in lesion-draining lymph nodes. Proteomic analysis confirmed regulation of a proinflammatory bone marrow response, which was abolished upon macrophage differentiation. Thus, in attenuation and regression of atherosclerosis, regardless of the monocyte signature, during monocyte to macrophage differentiation, proresolving macrophages prevail, mediating vascular repair. This study provides novel mechanistic insight into the monocyte/macrophage phenotypes in halted atherosclerosis progression and regression of atherosclerosis.-Bruen, R., Curley, S., Kajani, S., Lynch, G., O'Reilly, M. E., Dillon, E. T., Fitzsimons, S., Mthunzi, L., McGillicuddy, F. C., Belton, O. Different monocyte phenotypes result in proresolving macrophages in conjugated linoleic acid-induced attenuated progression and regression of atherosclerosis.
Collapse
Affiliation(s)
- Robyn Bruen
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Seán Curley
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Sarina Kajani
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Gina Lynch
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Marcella E O'Reilly
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Eugéne T Dillon
- Mass Spectrometry Resource, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Stephen Fitzsimons
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Liberty Mthunzi
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Fiona C McGillicuddy
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Orina Belton
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
26
|
Seiri P, Abi A, Soukhtanloo M. PPAR-γ: Its ligand and its regulation by microRNAs. J Cell Biochem 2019; 120:10893-10908. [PMID: 30770587 DOI: 10.1002/jcb.28419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/04/2018] [Indexed: 01/24/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor superfamily. PPARs are categorized into three subtypes, PPARα, β/δ, and γ, encoded by different genes, expressed in diverse tissues and participate in various biological functions and can be activated by their metabolic derivatives in the body or dietary fatty acids. The PPAR-γ also takes parts in the regulation of energy balance, lipoprotein metabolism, insulin sensitivity, oxidative stress, and inflammatory signaling. It has been implicated in the pathology of numerous diseases including obesity, diabetes, atherosclerosis, and cancers. Among various cellular and molecular targets that are able to regulate PPAR-γ and its underlying pathways, microRNAs (miRNAs) appeared as important regulators. Given that the deregulation of these molecules via targeting PPAR-γ could affect initiation and progression of various diseases, identification of miRNAs that affects PPAR-γ could contribute to the better understanding of roles of PPAR-γ in various biological and pathological conditions. Here, we have summarized the function and various ligands of PPAR-γ and have highlighted various miRNAs involved in the regulation of PPAR-γ.
Collapse
Affiliation(s)
- Parvaneh Seiri
- Department of Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Abi
- Department of Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
27
|
Bruen R, Fitzsimons S, Belton O. miR-155 in the Resolution of Atherosclerosis. Front Pharmacol 2019; 10:463. [PMID: 31139076 PMCID: PMC6527595 DOI: 10.3389/fphar.2019.00463] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/12/2019] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory disease where advanced lesions can eventually completely obstruct blood flow resulting in clinical events, such as a myocardial infarction or stroke. Monocytes and macrophages are the dominant biologically active immune cells involved in atherosclerosis disease and play a pivotal role during initiation, progression, and regression of disease. Altering macrophage inflammation is critical to induce regression of atherosclerosis and microRNAs (miRs) have emerged as key regulators of the macrophage phenotype. MiRs are small noncoding RNAs that regulate gene expression. They are dysregulated during atherosclerosis development and are key regulators of macrophage function and polarization. MiRs are short nucleotide transcripts that are very stable in circulation and thus have potential as therapeutics and/or biomarkers in the context of atherosclerosis. Of relevance to this review is that inhibition of macrophage-specific miR-155 may be a viable therapeutic strategy to decrease inflammation associated with atherosclerosis. However, further studies on these miRs and advancements in miR therapeutic delivery are required for these therapeutics to advance to the clinical setting. Conjugated linoleic acid (CLA), a pro-resolving lipid mediator, is an agonist of the peroxisome proliferator-activated receptor (PPAR)-γ. The biological activities of CLA have been documented to have anti-atherogenic effects in experimental models of atherosclerosis, inducing regression and impacting on monocyte and macrophage cells. Our work and that of others on PPAR-γ agonists and polyunsaturated fatty acids have shown that these mediators regulate candidate miRNAs and promote pro-resolving atherosclerotic plaque microenvironments.
Collapse
Affiliation(s)
- Robyn Bruen
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Stephen Fitzsimons
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Orina Belton
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
28
|
M2 Macrophages as a Potential Target for Antiatherosclerosis Treatment. Neural Plast 2019; 2019:6724903. [PMID: 30923552 PMCID: PMC6409015 DOI: 10.1155/2019/6724903] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammation course, which could induce life-threatening diseases such as stroke and myocardial infarction. Optimal medical treatments for atherosclerotic risk factors with current antihypertensive and lipid-lowering drugs (for example, statins) are widely used in clinical practice. However, many patients with established disease still continue to have recurrent cardiovascular events in spite of treatment with a state-of-the-art therapy. Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of mortality worldwide. Hence, current treatment of atherosclerosis is still far from being satisfactory. Recently, M2 macrophages have been found associated with atherosclerosis regression. The M2 phenotype can secrete anti-inflammatory factors such as IL-10 and TGF-β, promote tissue remodeling and repairing through collagen formation, and clear dying cells and debris by efferocytosis. Therefore, modulators targeting macrophages' polarization to the M2 phenotype could be another promising treatment strategy for atherosclerosis. Two main signaling pathways, the Akt/mTORC/LXR pathway and the JAK/STAT6 pathway, are found playing important roles in M2 polarization. In addition, researchers have reported several potential approaches to modulate M2 polarization. Inhibiting or activating some kinds of enzymes, affecting transcription factors, or acting on several membrane receptors could regulate the polarization of the M2 phenotype. Besides, biomolecules, for example vitamin D, were found to affect the process of M2 polarization. Pomegranate juice could promote M2 polarization via unclear mechanism. In this review, we will discuss how M2 macrophages affect atherosclerosis regression, signal transduction in M2 polarization, and outline potential targets and compounds that affect M2 polarization, thus controlling the progress of atherosclerosis.
Collapse
|
29
|
Fakan B, Szalardy L, Vecsei L. Exploiting the Therapeutic Potential of Endogenous Immunomodulatory Systems in Multiple Sclerosis-Special Focus on the Peroxisome Proliferator-Activated Receptors (PPARs) and the Kynurenines. Int J Mol Sci 2019; 20:ijms20020426. [PMID: 30669473 PMCID: PMC6358998 DOI: 10.3390/ijms20020426] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/13/2019] [Accepted: 01/15/2019] [Indexed: 01/20/2023] Open
Abstract
Multiple sclerosis (MS) is a progressive neurodegenerative disease, characterized by autoimmune central nervous system (CNS) demyelination attributable to a disturbed balance between encephalitic T helper 1 (Th1) and T helper 17 (Th17) and immunomodulatory regulatory T cell (Treg) and T helper 2 (Th2) cells, and an alternatively activated macrophage (M2) excess. Endogenous molecular systems regulating these inflammatory processes have recently been investigated to identify molecules that can potentially influence the course of the disease. These include the peroxisome proliferator-activated receptors (PPARs), PPARγ coactivator-1alpha (PGC-1α), and kynurenine pathway metabolites. Although all PPARs ameliorate experimental autoimmune encephalomyelitis (EAE), recent evidence suggests that PPARα, PPARβ/δ agonists have less pronounced immunomodulatory effects and, along with PGC-1α, are not biomarkers of neuroinflammation in contrast to PPARγ. Small clinical trials with PPARγ agonists have been published with positive results. Proposed as immunomodulatory and neuroprotective, the therapeutic use of PGC-1α activation needs to be assessed in EAE/MS. The activation of indolamine 2,3-dioxygenase (IDO), the rate-limiting step of the kynurenine pathway of tryptophan (Trp) metabolism, plays crucial immunomodulatory roles. Indeed, Trp metabolites have therapeutic relevance in EAE and drugs with structural analogy to kynurenines, such as teriflunomide, are already approved for MS. Further studies are required to gain deeper knowledge of such endogenous immunomodulatory pathways with potential therapeutic implications in MS.
Collapse
Affiliation(s)
- Bernadett Fakan
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Semmelweis u. 6, Hungary.
| | - Levente Szalardy
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Semmelweis u. 6, Hungary.
| | - Laszlo Vecsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Semmelweis u. 6, Hungary.
- MTA-SZTE Neuroscience Research Group, H-6725 Szeged, Semmelweis u. 6, Hungary.
| |
Collapse
|
30
|
Baal N, Cunningham S, Obermann HL, Thomas J, Lippitsch A, Dietert K, Gruber AD, Kaufmann A, Michel G, Nist A, Stiewe T, Rupp O, Goesmann A, Zukunft S, Fleming I, Bein G, Lohmeyer J, Bauer S, Hackstein H. ADAR1 Is Required for Dendritic Cell Subset Homeostasis and Alveolar Macrophage Function. THE JOURNAL OF IMMUNOLOGY 2019; 202:1099-1111. [DOI: 10.4049/jimmunol.1800269] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 12/07/2018] [Indexed: 11/19/2022]
|
31
|
Regulation of macrophage immunometabolism in atherosclerosis. Nat Immunol 2018; 19:526-537. [PMID: 29777212 DOI: 10.1038/s41590-018-0113-3] [Citation(s) in RCA: 386] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023]
Abstract
After activation, cells of the myeloid lineage undergo robust metabolic transitions, as well as discrete epigenetic changes, that can dictate both ongoing and future inflammatory responses. In atherosclerosis, in which macrophages play central roles in the initiation, growth, and ultimately rupture of arterial plaques, altered metabolism is a key feature that dictates macrophage function and subsequent disease progression. This Review explores how factors central to the plaque microenvironment (for example, altered cholesterol metabolism, oxidative stress, hypoxia, apoptotic and necrotic cells, and hyperglycemia) shape the metabolic rewiring of macrophages in atherosclerosis as well as how these metabolic shifts in turn alter macrophage immune-effector and tissue-reparative functions. Finally, this overview offers insight into the challenges and opportunities of harnessing metabolism to modulate aberrant macrophage responses in disease.
Collapse
|
32
|
Zhao LJ, Zhang SF. Activation of TGR5 promotes mitochondrial biogenesis in human aortic endothelial cells. Biochem Biophys Res Commun 2018; 500:952-957. [PMID: 29709472 DOI: 10.1016/j.bbrc.2018.04.210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 01/22/2023]
Abstract
Impairment of mitochondrial biogenesis has been associated with vascular pathophysiology. The G-protein-coupled receptor (TGR5) is an important mediator of bile acid signaling and glucose metabolism. However, the effects of TGR5 on mitochondrial biogenesis in endothelial cells remain elusive. In this study, we found that activation of TGR5 using its specific agonist taurolithocholic acid (TLCA) promoted the expression of PGC-1α, a master regulator of mitochondrial biogenesis in human aortic endothelial cells (HAECs). Additionally, activation of TGR5 increased the expression of PGC-1α target genes, such as NRF1 and TFAM. Indeed, we found that TLCA treatment promoted mitochondrial biogenesis by increasing mitochondrial mass, mitochondrial-to-nuclear DNA (mtDNA/nDNA), COX-Ⅰ expression, and cytochrome c oxidase activity in HAECs. Notably, our results displayed that activation of TGR5 resulted in a functional gain in mitochondria by increasing the rate of respiration and ATP production. Mechanistically, we found that TLCA treatment activated the transcriptional factor CREB by inducing the phosphorylation of CREB at Ser133. Using the PKA/CREB inhibitor H89 abolished the effects of TLCA on PGC-1α, NRF1 and TFAM expression as well as the increase in mtDNA/nDNA and ATP production. These findings suggest that activation of TGR5 promoted mitochondrial biogenesis in endothelial cells, which is mediated by the CREB/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Li-Jun Zhao
- Department of Internal Medicine, West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China.
| | - Shi-Fang Zhang
- Department of Pulmonary Disease, Institute of Respiratory Disease, Chengdu Second People's Hospital, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
33
|
Vozenilek AE, Navratil AR, Green JM, Coleman DT, Blackburn CMR, Finney AC, Pearson BH, Chrast R, Finck BN, Klein RL, Orr AW, Woolard MD. Macrophage-Associated Lipin-1 Enzymatic Activity Contributes to Modified Low-Density Lipoprotein-Induced Proinflammatory Signaling and Atherosclerosis. Arterioscler Thromb Vasc Biol 2017; 38:324-334. [PMID: 29217509 DOI: 10.1161/atvbaha.117.310455] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Macrophage proinflammatory responses induced by modified low-density lipoproteins (modLDL) contribute to atherosclerotic progression. How modLDL causes macrophages to become proinflammatory is still enigmatic. Macrophage foam cell formation induced by modLDL requires glycerolipid synthesis. Lipin-1, a key enzyme in the glycerolipid synthesis pathway, contributes to modLDL-elicited macrophage proinflammatory responses in vitro. The objective of this study was to determine whether macrophage-associated lipin-1 contributes to atherogenesis and to assess its role in modLDL-mediated signaling in macrophages. APPROACH AND RESULTS We developed mice lacking lipin-1 in myeloid-derived cells and used adeno-associated viral vector 8 expressing the gain-of-function mutation of mouse proprotein convertase subtilisin/kexin type 9 (adeno-associated viral vector 8-proprotein convertase subtilisin/kexin type 9) to induce hypercholesterolemia and plaque formation. Mice lacking myeloid-associated lipin-1 had reduced atherosclerotic burden compared with control mice despite similar plasma lipid levels. Stimulation of bone marrow-derived macrophages with modLDL activated a persistent protein kinase Cα/βII-extracellular receptor kinase1/2-jun proto-oncogene signaling cascade that contributed to macrophage proinflammatory responses that was dependent on lipin-1 enzymatic activity. CONCLUSIONS Our data demonstrate that macrophage-associated lipin-1 is atherogenic, likely through persistent activation of a protein kinase Cα/βII-extracellular receptor kinase1/2-jun proto-oncogene signaling cascade that contributes to foam cell proinflammatory responses. Taken together, these results suggest that modLDL-induced foam cell formation and modLDL-induced macrophage proinflammatory responses are not independent consequences of modLDL stimulation but rather are both directly influenced by enhanced lipid synthesis.
Collapse
Affiliation(s)
- Aimee E Vozenilek
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Aaron R Navratil
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Jonette M Green
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - David T Coleman
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Cassidy M R Blackburn
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Alexandra C Finney
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Brenna H Pearson
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Roman Chrast
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Brian N Finck
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Ronald L Klein
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - A Wayne Orr
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.)
| | - Matthew D Woolard
- From the Department of Microbiology and Immunology (A.E.V., C.M.R.B., M.D.W.), Department of Pathology and Translational Pathobiology (J.M.G., B.H.P., A.W.O.), Department of Cell Biology and Anatomy (A.C.F.), Feist-Weiller Cancer Center (D.T.C.), and Pharmacology, Toxicology, and Neuroscience (R.L.K.), Louisiana State University Health Sciences Center, Shreveport; Department of Pharmacology, University of California San Diego, La Jolla (A.R.N.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (R.C.); and Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO (B.N.F.).
| |
Collapse
|
34
|
Vilne B, Skogsberg J, Foroughi Asl H, Talukdar HA, Kessler T, Björkegren JLM, Schunkert H. Network analysis reveals a causal role of mitochondrial gene activity in atherosclerotic lesion formation. Atherosclerosis 2017; 267:39-48. [PMID: 29100060 DOI: 10.1016/j.atherosclerosis.2017.10.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/05/2017] [Accepted: 10/18/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS Mitochondrial damage and augmented production of reactive oxygen species (ROS) may represent an intermediate step by which hypercholesterolemia exacerbates atherosclerotic lesion formation. METHODS To test this hypothesis, in mice with severe but genetically reversible hypercholesterolemia (i.e. the so called Reversa mouse model), we performed time-resolved analyses of mitochondrial transcriptome in the aortic arch employing a systems-level network approach. RESULTS During hypercholesterolemia, we observed a massive down-regulation (>28%) of mitochondrial genes, specifically at the time of rapid atherosclerotic lesion expansion and foam cell formation, i.e. between 30 and 40 weeks of age. Both phenomena - down-regulation of mitochondrial genes and lesion expansion - were largely reversible by genetically lowering plasma cholesterol (by >80%, from 427 to 54 ± 31 mg/L) at 30 weeks. Co-expression network analysis revealed that both mitochondrial signature genes were highly connected in two modules, negatively correlating with lesion size and supported as causal for coronary artery disease (CAD) in humans, as expression-associated single nucleotide polymorphisms (eSNPs) representing their genes overlapped markedly with established disease risk loci. Within these modules, we identified the transcription factor estrogen related receptor (ERR)-α and its co-factors PGC1-α and -β, i.e. two members of the peroxisome proliferator-activated receptor γ co-activator 1 family of transcription regulators, as key regulatory genes. Together, these factors are known as major orchestrators of mitochondrial biogenesis and antioxidant responses. CONCLUSIONS Using a network approach, we demonstrate how hypercholesterolemia could hamper mitochondrial activity during atherosclerosis progression and pinpoint potential therapeutic targets to counteract these processes.
Collapse
Affiliation(s)
- Baiba Vilne
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; DZHK (German Research Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Josefin Skogsberg
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hassan Foroughi Asl
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Husain Ahammad Talukdar
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; DZHK (German Research Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Johan L M Björkegren
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Estonia; Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai New York, New York, USA; Clinical Gene Networks AB, Stockholm, Sweden.
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; DZHK (German Research Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
35
|
Silveira LS, Pimentel GD, Souza CO, Biondo LA, Teixeira AAS, Lima EA, Batatinha HAP, Rosa Neto JC, Lira FS. Effect of an acute moderate-exercise session on metabolic and inflammatory profile of PPAR-α knockout mice. Cell Biochem Funct 2017; 35:510-517. [DOI: 10.1002/cbf.3308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/23/2017] [Accepted: 09/17/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Loreana S. Silveira
- Exercise and Immunometabolism Research Group, Department of Physical Education; Universidade Estadual Paulista; Presidente Prudente SP Brazil
| | - Gustavo D. Pimentel
- Clinical and Sports Nutrition Research Laboratory (Labince); Nutrition Faculty (FANUT)-Federal University of Goiás (UFG); Goiânia GO Brazil
| | - Camila O. Souza
- Immunometabolism Research Group, Institute of Biomedical Sciences; University of São Paulo (USP); São Paulo SP Brazil
| | - Luana A. Biondo
- Immunometabolism Research Group, Institute of Biomedical Sciences; University of São Paulo (USP); São Paulo SP Brazil
| | - Alexandre Abílio S. Teixeira
- Immunometabolism Research Group, Institute of Biomedical Sciences; University of São Paulo (USP); São Paulo SP Brazil
| | - Edson A. Lima
- Immunometabolism Research Group, Institute of Biomedical Sciences; University of São Paulo (USP); São Paulo SP Brazil
| | - Helena A. P. Batatinha
- Immunometabolism Research Group, Institute of Biomedical Sciences; University of São Paulo (USP); São Paulo SP Brazil
| | - José C. Rosa Neto
- Immunometabolism Research Group, Institute of Biomedical Sciences; University of São Paulo (USP); São Paulo SP Brazil
| | - Fábio S. Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education; Universidade Estadual Paulista; Presidente Prudente SP Brazil
| |
Collapse
|
36
|
Song X, Yang B, Qiu F, Jia M, Fu G. High glucose and free fatty acids induce endothelial progenitor cell senescence via PGC-1α/SIRT1 signaling pathway. Cell Biol Int 2017; 41:1146-1159. [PMID: 28786152 DOI: 10.1002/cbin.10833] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/28/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Xiaoxiao Song
- The Department of Endocrinology; Second Affiliated Hospital, College of Medicine, Zhejiang University; Hangzhou 310009 Zhejiang Province China
- The Department of Cardiology; Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University; Hangzhou 310016 Zhejiang Province China
| | - Boyun Yang
- The Department of Endocrinology; Second Affiliated Hospital, College of Medicine, Zhejiang University; Hangzhou 310009 Zhejiang Province China
| | - Fuyu Qiu
- The Department of Cardiology; Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University; Hangzhou 310016 Zhejiang Province China
| | - Minyue Jia
- The Department of Endocrinology; Second Affiliated Hospital, College of Medicine, Zhejiang University; Hangzhou 310009 Zhejiang Province China
| | - Guosheng Fu
- The Department of Cardiology; Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University; Hangzhou 310016 Zhejiang Province China
| |
Collapse
|
37
|
Kadlec AO, Chabowski DS, Ait-Aissa K, Hockenberry JC, Otterson MF, Durand MJ, Freed JK, Beyer AM, Gutterman DD. PGC-1α (Peroxisome Proliferator-Activated Receptor γ Coactivator 1-α) Overexpression in Coronary Artery Disease Recruits NO and Hydrogen Peroxide During Flow-Mediated Dilation and Protects Against Increased Intraluminal Pressure. Hypertension 2017; 70:166-173. [PMID: 28533333 DOI: 10.1161/hypertensionaha.117.09289] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/12/2017] [Accepted: 04/20/2017] [Indexed: 12/27/2022]
Abstract
Blood flow through healthy human vessels releases NO to produce vasodilation, whereas in patients with coronary artery disease (CAD), the mediator of dilation transitions to mitochondria-derived hydrogen peroxide (mtH2O2). Excessive mtH2O2 production contributes to a proatherosclerotic vascular milieu. Loss of PGC-1α (peroxisome proliferator-activated receptor γ coactivator 1α) is implicated in the pathogenesis of CAD. We hypothesized that PGC-1α suppresses mtH2O2 production to reestablish NO-mediated dilation in isolated vessels from patients with CAD. Isolated human adipose arterioles were cannulated, and changes in lumen diameter in response to graded increases in flow were recorded in the presence of PEG (polyethylene glycol)-catalase (H2O2 scavenger) or L-NAME (NG-nitro-l-arginine methyl ester; NOS inhibitor). In contrast to the exclusively NO- or H2O2-mediated dilation seen in either non-CAD or CAD conditions, respectively, flow-mediated dilation in CAD vessels was sensitive to both L-NAME and PEG-catalase after PGC-1α upregulation using ZLN005 and α-lipoic acid. PGC-1α overexpression in CAD vessels protected against the vascular dysfunction induced by an acute increase in intraluminal pressure. In contrast, downregulation of PGC-1α in non-CAD vessels produces a CAD-like phenotype characterized by mtH2O2-mediated dilation (no contribution of NO). Loss of PGC-1α may contribute to the shift toward the mtH2O2-mediated dilation observed in vessels from subjects with CAD. Strategies to boost PGC-1α levels may provide a therapeutic option in patients with CAD by shifting away from mtH2O2-mediated dilation, increasing NO bioavailability, and reducing levels of mtH2O2 Furthermore, increased expression of PGC-1α allows for simultaneous contributions of both NO and H2O2 to flow-mediated dilation.
Collapse
Affiliation(s)
- Andrew O Kadlec
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Dawid S Chabowski
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Karima Ait-Aissa
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Joseph C Hockenberry
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Mary F Otterson
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Matthew J Durand
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Julie K Freed
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - Andreas M Beyer
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee
| | - David D Gutterman
- From the Department of Physiology (A.O.K., A.M.B., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., J.C.H., A.M.B., D.D.G.), Cardiovascular Center (A.O.K., D.S.C., K.A.-A., J.C.H., M.J.D., J.K.F., A.M.B., D.D.G.), Department of Physical Medicine and Rehabilitation (M.J.D.), Division of Colorectal Surgery, Department of Surgery (M.F.O.), and Department of Anesthesiology (J.K.F.), Medical College of Wisconsin, Milwaukee.
| |
Collapse
|
38
|
Balci Yuce H, Akbulut N, Ocakli S, Kayir O, Elmastas M. The effect of commercial conjugated linoleic acid products on experimental periodontitis and diabetes mellitus in Wistar rats. Acta Odontol Scand 2017; 75:21-29. [PMID: 27897090 DOI: 10.1080/00016357.2016.1244355] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The aim of present study was to determine the effects of conjugated linoleic acid enriched milk on alveolar bone loss, hyperglycaemia, oxidative stress and apoptosis in ligature-induced periodontal disease in diabetic rat model. METHODS Wistar rats were divided into six experimental groups: 1; non-ligated (NL, n = 6) group, 2; ligature only (LO, n = 6) group, 3; streptozotocin only (STZ, n = 8) group, 4; STZ and ligature (STZ + L, n = 8) group, 5; ligature and conjugated linoleic acid (CLA) (L + CLA, n = 8) group, 6; STZ, ligature and CLA group (STZ + L + CLA, n = 8) group. Diabetes mellitus was induced by 60 mg/kg streptozotocin. Rats were fed with CLA enriched milk for four weeks. Silk ligatures were placed at the gingival margin of lower first molars of mandibular quadrant. The study duration was four weeks after diabetes induction and the animals were sacrificed at the end of this period. Changes in alveolar bone levels were clinically measured and tissues were histopathologically examined. Inducible nitric oxide synthase (iNOS) and Bax protein expressions, serum interleukin-1β (IL-1β), low-density lipoprotein (LDL), high-density lipoprotein (HDL) and triglyceride levels and tartrate resistant acid phosphatase (TRAP)+ osteoclast numbers were also evaluated. RESULTS At the end of four weeks, alveolar bone loss was significantly higher in the STZ + LO group compared to the other groups (p < .05). CLA decreased alveolar bone loss in L + CLA and STZ + L + CLA groups. CLA significantly decreased TRAP + osteoclast numbers and increased osteoblastic activity compared to the STZ + L group (p < .05). Diabetes and CLA increased Bax protein levels (p < .05) however CLA had no effect on iNOS expression (p > .05). CONCLUSION Within the limits of this study, commercial CLA product administration in addition to diet significantly reduced alveolar bone loss, increased osteoblastic activity and decreased osteoclastic activity in the diabetic Wistar rats.
Collapse
|
39
|
de Gaetano M, Crean D, Barry M, Belton O. M1- and M2-Type Macrophage Responses Are Predictive of Adverse Outcomes in Human Atherosclerosis. Front Immunol 2016; 7:275. [PMID: 27486460 PMCID: PMC4949256 DOI: 10.3389/fimmu.2016.00275] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/01/2016] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis is an inflammatory disease caused by endothelial injury, lipid deposition, and oxidative stress. This progressive disease can be converted into an acute clinical event by plaque rupture and thrombosis. In the context of atherosclerosis, the underlying cause of myocardial infarction and stroke, macrophages uniquely possess a dual functionality, regulating lipid accumulation and metabolism and sustaining the chronic inflammatory response, two of the most well-documented pathways associated with the pathogenesis of the disease. Macrophages are heterogeneous cell populations and it is hypothesized that, during the pathogenesis of atherosclerosis, macrophages in the developing plaque can switch from a pro-inflammatory (MΦ1) to an anti-inflammatory (MΦ2) phenotype and vice versa, depending on the microenvironment. The aim of this study was to identify changes in macrophage subpopulations in the progression of human atherosclerotic disease. Established atherosclerotic plaques from symptomatic and asymptomatic patients with existing coronary artery disease undergoing carotid endarterectomy were recruited to the study. Comprehensive histological and immunohistochemical analyses were performed to quantify the cellular content and macrophage subsets of atherosclerotic lesion. In parallel, expression of MΦ1 and MΦ2 macrophage markers were analyzed by real-time PCR and Western blot analysis. Gross analysis and histological staining demonstrated that symptomatic plaques presented greater hemorrhagic activity and the internal carotid was the most diseased segment, based on the predominant prevalence of fibrotic and necrotic tissue, calcifications, and hemorrhagic events. Immunohistochemical analysis showed that both MΦ1 and MΦ2 macrophages are present in human plaques. However, MΦ2 macrophages are localized to more stable locations within the lesion. Importantly, gene and protein expression analysis of MΦ1/MΦ2 markers evidenced that MΦ1 markers and Th1-associated cytokines are highly expressed in symptomatic plaques, whereas expression of the MΦ2 markers, mannose receptor (MR), and CD163 and Th2 cytokines are inversely related with disease progression. These data increase the understanding of atherosclerosis development, identifying the cellular content of lesions during disease progression, and characterizing macrophage subpopulation within human atherosclerotic plaques.
Collapse
Affiliation(s)
- Monica de Gaetano
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin , Ireland
| | - Daniel Crean
- School of Veterinary Medicine, UCD Conway Institute, University College Dublin , Dublin , Ireland
| | - Mary Barry
- St. Vincent's University Hospital , Dublin , Ireland
| | - Orina Belton
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin , Ireland
| |
Collapse
|
40
|
Kadlec AO, Chabowski DS, Ait-Aissa K, Gutterman DD. Role of PGC-1α in Vascular Regulation: Implications for Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:1467-74. [PMID: 27312223 DOI: 10.1161/atvbaha.116.307123] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/02/2016] [Indexed: 11/16/2022]
Abstract
Mitochondrial dysfunction results in high levels of oxidative stress and mitochondrial damage, leading to disruption of endothelial homeostasis. Recent discoveries have clarified several pathways, whereby mitochondrial dysregulation contributes to endothelial dysfunction and vascular disease burden. One such pathway centers around peroxisome proliferator receptor-γ coactivator 1α (PGC-1α), a transcriptional coactivator linked to mitochondrial biogenesis and antioxidant defense, among other functions. Although primarily investigated for its therapeutic potential in obesity and skeletal muscle differentiation, the ability of PGC-1α to alter a multitude of cellular functions has sparked interest in its role in the vasculature. Within this context, recent studies demonstrate that PGC-1α plays a key role in endothelial cell and smooth muscle cell regulation through effects on oxidative stress, apoptosis, inflammation, and cell proliferation. The ability of PGC-1α to affect these parameters is relevant to vascular disease progression, particularly in relation to atherosclerosis. Upregulation of PGC-1α can prevent the development of, and even encourage regression of, atherosclerotic lesions. Therefore, PGC-1α is poised to serve as a promising target in vascular disease. This review details recent findings related to PGC-1α in vascular regulation, regulation of PGC-1α itself, the role of PGC-1α in atherosclerosis, and therapies that target this key protein.
Collapse
Affiliation(s)
- Andrew O Kadlec
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.)
| | - Dawid S Chabowski
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.)
| | - Karima Ait-Aissa
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.)
| | - David D Gutterman
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.).
| |
Collapse
|
41
|
Bruen R, Fitzsimons S, Belton O. Atheroprotective effects of conjugated linoleic acid. Br J Clin Pharmacol 2016; 83:46-53. [PMID: 27037767 DOI: 10.1111/bcp.12948] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/22/2016] [Accepted: 03/28/2016] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis, the underlying cause of heart attack and strokes, is a progressive dyslipidaemic and inflammatory disease where monocyte-derived macrophage cells play a pivotal role. Although most of the mechanisms that contribute to the progression of atherosclerosis have been identified, there is limited information on those governing regression. Conjugated linoleic acid (CLA) is a generic term denoting a group of naturally occurring isomers of linoleic acid (18:2, n6) that differ in the position or geometry (i.e. cis or trans) of their double bonds. The most predominant isomers in ruminant fats are cis-9, trans-11 CLA (c9,t11-CLA), which accounts for more than 80% of CLA isomers in dairy products and trans-10, cis-12 CLA (t10,c12-CLA). Dietary administration of a blend of the two most abundant isomers of CLA has been shown to inhibit the progression and induce the regression of pre-established atherosclerosis. Studies investigating the mechanisms involved in CLA-induced atheroprotective effects are continually emerging. The purpose of this review is to discuss comprehensively the effects of CLA on monocyte/macrophage function in atherosclerosis and to identify possible mechanisms through which CLA mediates its atheroprotective effects.
Collapse
Affiliation(s)
- Robyn Bruen
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Ireland
| | - Stephen Fitzsimons
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Ireland
| | - Orina Belton
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Ireland
| |
Collapse
|
42
|
Abstract
Nuclear receptors sense a wide range of steroids and hormones (estrogens, progesterone, androgens, glucocorticoid, and mineralocorticoid), vitamins (A and D), lipid metabolites, carbohydrates, and xenobiotics. In response to these diverse but critically important mediators, nuclear receptors regulate the homeostatic control of lipids, carbohydrate, cholesterol, and xenobiotic drug metabolism, inflammation, cell differentiation and development, including vascular development. The nuclear receptor family is one of the most important groups of signaling molecules in the body and as such represent some of the most important established and emerging clinical and therapeutic targets. This review will highlight some of the recent trends in nuclear receptor biology related to vascular biology.
Collapse
Affiliation(s)
- David Bishop-Bailey
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK,
| |
Collapse
|
43
|
Xiu F, Diao L, Qi P, Catapano M, Jeschke MG. Palmitate differentially regulates the polarization of differentiating and differentiated macrophages. Immunology 2015; 147:82-96. [PMID: 26453839 DOI: 10.1111/imm.12543] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 08/31/2015] [Accepted: 09/17/2015] [Indexed: 12/27/2022] Open
Abstract
The tissue accumulation of M1 macrophages in patients with metabolic diseases such as obesity and type 2 diabetes mellitus has been well-documented. Interestingly, it is an accumulation of M2 macrophages that is observed in the adipose, liver and lung tissues, as well as in the circulation, of patients who have had major traumas such as a burn injury or sepsis; however, the trigger for the M2 polarization observed in these patients has not yet been identified. In the current study, we explored the effects of chronic palmitate and high glucose treatment on macrophage differentiation and function in murine bone-marrow-derived macrophages. We found that chronic treatment with palmitate decreased phagocytosis and HLA-DR expression in addition to inhibiting the production of pro-inflammatory cytokines. Chronic palmitate treatment of bone marrows also led to M2 polarization, which correlated with the activation of the peroxisome proliferator-activated receptor-γ signalling pathway. Furthermore, we found that chronic palmitate treatment increased the expression of multiple endoplasmic reticulum (ER) stress markers, including binding immunoglobulin protein. Preconditioning with the universal ER stress inhibitor 4-phenylbutyrate attenuated ER stress signalling and neutralized the effect of palmitate, inducing a pro-inflammatory phenotype. We confirmed these results in differentiating human macrophages, showing an anti-inflammatory response to chronic palmitate exposure. Though alone it did not promote M2 polarization, hyperglycaemia exacerbated the effects of palmitate. These findings suggest that the dominant accumulation of M2 in adipose tissue and liver in patients with critical illness may be a result of hyperlipidaemia and hyperglycaemia, both components of the hypermetabolism observed in critically ill patients.
Collapse
Affiliation(s)
- Fangming Xiu
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Li Diao
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Peter Qi
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Michael Catapano
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Marc G Jeschke
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.,Division of Plastic Surgery, Department of Surgery, Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
Xiong S, Patrushev N, Forouzandeh F, Hilenski L, Alexander RW. PGC-1α Modulates Telomere Function and DNA Damage in Protecting against Aging-Related Chronic Diseases. Cell Rep 2015; 12:1391-9. [PMID: 26299964 DOI: 10.1016/j.celrep.2015.07.047] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/18/2015] [Accepted: 07/23/2015] [Indexed: 12/24/2022] Open
Abstract
Cellular senescence and organismal aging predispose age-related chronic diseases, such as neurodegenerative, metabolic, and cardiovascular disorders. These diseases emerge coincidently from elevated oxidative/electrophilic stress, inflammation, mitochondrial dysfunction, DNA damage, and telomere dysfunction and shortening. Mechanistic linkages are incompletely understood. Here, we show that ablation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) accelerates vascular aging and atherosclerosis, coinciding with telomere dysfunction and shortening and DNA damage. PGC-1α deletion reduces expression and activity of telomerase reverse transcriptase (TERT) and increases p53 levels. Ectopic expression of PGC-1α coactivates TERT transcription and reverses telomere malfunction and DNA damage. Furthermore, alpha lipoic acid (ALA), a non-dispensable mitochondrial cofactor, upregulates PGC-1α-dependent TERT and the cytoprotective Nrf-2-mediated antioxidant/electrophile-responsive element (ARE/ERE) signaling cascades, and counteracts high-fat-diet-induced, age-dependent arteriopathy. These results illustrate the pivotal importance of PGC-1α in ameliorating senescence, aging, and associated chronic diseases, and may inform novel therapeutic approaches involving electrophilic specificity.
Collapse
Affiliation(s)
- Shiqin Xiong
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nikolay Patrushev
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Farshad Forouzandeh
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lula Hilenski
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - R Wayne Alexander
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
45
|
Zhang Z, Guo M, Zhao S, Xu W, Shao J, Zhang F, Wu L, Lu Y, Zheng S. The update on transcriptional regulation of autophagy in normal and pathologic cells: A novel therapeutic target. Biomed Pharmacother 2015; 74:17-29. [DOI: 10.1016/j.biopha.2015.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/15/2015] [Indexed: 02/08/2023] Open
|
46
|
Karunakaran D, Thrush AB, Nguyen MA, Richards L, Geoffrion M, Singaravelu R, Ramphos E, Shangari P, Ouimet M, Pezacki JP, Moore KJ, Perisic L, Maegdefessel L, Hedin U, Harper ME, Rayner KJ. Macrophage Mitochondrial Energy Status Regulates Cholesterol Efflux and Is Enhanced by Anti-miR33 in Atherosclerosis. Circ Res 2015; 117:266-78. [PMID: 26002865 DOI: 10.1161/circresaha.117.305624] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 05/22/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE Therapeutically targeting macrophage reverse cholesterol transport is a promising approach to treat atherosclerosis. Macrophage energy metabolism can significantly influence macrophage phenotype, but how this is controlled in foam cells is not known. Bioinformatic pathway analysis predicts that miR-33 represses a cluster of genes controlling cellular energy metabolism that may be important in macrophage cholesterol efflux. OBJECTIVE We hypothesized that cellular energy status can influence cholesterol efflux from macrophages, and that miR-33 reduces cholesterol efflux via repression of mitochondrial energy metabolism pathways. METHODS AND RESULTS In this study, we demonstrated that macrophage cholesterol efflux is regulated by mitochondrial ATP production, and that miR-33 controls a network of genes that synchronize mitochondrial function. Inhibition of mitochondrial ATP synthase markedly reduces macrophage cholesterol efflux capacity, and anti-miR33 required fully functional mitochondria to enhance ABCA1-mediated cholesterol efflux. Specifically, anti-miR33 derepressed the novel target genes PGC-1α, PDK4, and SLC25A25 and boosted mitochondrial respiration and production of ATP. Treatment of atherosclerotic Apoe(-/-) mice with anti-miR33 oligonucleotides reduced aortic sinus lesion area compared with controls, despite no changes in high-density lipoprotein cholesterol or other circulating lipids. Expression of miR-33a/b was markedly increased in human carotid atherosclerotic plaques compared with normal arteries, and there was a concomitant decrease in mitochondrial regulatory genes PGC-1α, SLC25A25, NRF1, and TFAM, suggesting these genes are associated with advanced atherosclerosis in humans. CONCLUSIONS This study demonstrates that anti-miR33 therapy derepresses genes that enhance mitochondrial respiration and ATP production, which in conjunction with increased ABCA1 expression, works to promote macrophage cholesterol efflux and reduce atherosclerosis.
Collapse
Affiliation(s)
- Denuja Karunakaran
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - A Brianne Thrush
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - My-Anh Nguyen
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Laura Richards
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Michele Geoffrion
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Ragunath Singaravelu
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Eleni Ramphos
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Prakriti Shangari
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Mireille Ouimet
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - John P Pezacki
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Kathryn J Moore
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Ljubica Perisic
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Lars Maegdefessel
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Ulf Hedin
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Mary-Ellen Harper
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden
| | - Katey J Rayner
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (D.K., M.-A.N., L.R., M.G., E.R., P.S., K.J.R.); Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada (A.B.T., M.-A.N., R.S., J.P.P., M.-E.H., K.J.R.); National Research Council of Canada, Ottawa, Ontario, Canada (R.S., J.P.P.); Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine (M.O., K.J.M.); and Department of Molecular Medicine and Surgery (L.P., U.H.) and Department of Medicine (L.M.), Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
47
|
de Gaetano M, Alghamdi K, Marcone S, Belton O. Conjugated linoleic acid induces an atheroprotective macrophage MΦ2 phenotype and limits foam cell formation. JOURNAL OF INFLAMMATION-LONDON 2015; 12:15. [PMID: 25722654 PMCID: PMC4340802 DOI: 10.1186/s12950-015-0060-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/03/2015] [Indexed: 01/01/2023]
Abstract
Background Atherosclerosis, the underlying cause of heart attack and strokes, is a progresive dyslipidemic and inflammatory disease where monocyte-derived macrophage cells play a pivotal role. Although most of the mechanisms that contribute to the progression of atherosclerosis have been identified, there is limited information on those governing regression. Conjugated linoleic acid (CLA) is a group of isomers of linoleic acid that differ in the position and/or geometry of their double bonds. We have previously shown that a specific CLA blend (80:20 cis-9,trans-11:trans-10,cis-12-CLA) induces regression of pre-established atherosclerosis in vivo, via modulation of monocyte/macrophage function. However, the exact mechanisms through which CLA mediates this effect remain to be elucidated. Methods Here, we address if CLA primes monocytes towards an anti-inflammatory MΦ2 macrophage and examine the effect of individual CLA isomers and the atheroprotective blend on monocyte-macrophage differentiation, cytokine generation, foam cell formation and cholesterol metabolism in human peripheral blood monocyte (HPBMC)-derived macrophages. Results cis-9,trans-11-CLA and the atheroprotective 80:20 CLA blend regulates expression of pro-inflammatory mediators and modulates the inflammatory cytokine profile of macrophages and foam cells. In addition, cis-9,trans-11-CLA and CLA blend primes HPBMCs towards an anti-inflammatory MΦ2 phenotype, characterised by increased scavenger receptor (CD36) and efflux protein (ABCA-1) expression. Furthermore, this altered macrophage phenotype impacts on foam cell formation, inhibiting ox-LDL accumulation and promoting cholesterol efflux via both PPARγ and LXRα dependent pathways. Conclusion The data increases the understanding of the pathways regulated by CLA in atheroprotection, namely, inhibiting the progressive acquisition of a pro-inflammatory macrophage phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s12950-015-0060-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Monica de Gaetano
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Kawthar Alghamdi
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Simone Marcone
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Orina Belton
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
48
|
Zhu L, Zhao Q, Yang T, Ding W, Zhao Y. Cellular metabolism and macrophage functional polarization. Int Rev Immunol 2014; 34:82-100. [PMID: 25340307 DOI: 10.3109/08830185.2014.969421] [Citation(s) in RCA: 302] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Macrophages are a functionally heterogeneous cell population that is mainly shaped by a variety of microenvironmental stimuli. Interferon γ (IFN-γ), interleukin-1β (IL-1β), and lipopolysaccharide (LPS) induce a classical activation of macrophages (M1), whereas IL-4 and IL-13 induce an alternative activation program in macrophages (M2). Reprogramming of intracellular metabolisms is required for the proper polarization and functions of activated macrophages. Similar to the Warburg effect observed in tumor cells, M1 macrophages increase glucose consumption and lactate release and decreased oxygen consumption rate. In comparison, M2 macrophages mainly employ oxidative glucose metabolism pathways. In addition, fatty acids, vitamins, and iron metabolisms are also related to macrophage polarization. However, detailed metabolic pathways involved in macrophages have remained elusive. Understanding the bidirectional interactions between cellular metabolism and macrophage functions in physiological and pathological situations and the regulatory pathways involved may offer novel therapies for macrophage-associated diseases.
Collapse
Affiliation(s)
- Linnan Zhu
- 1Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | |
Collapse
|
49
|
Piell KM, Qipshidze Kelm N, Caroway MP, Aman M, Cole MP. Nitrite treatment rescues cardiac dysfunction in aged mice treated with conjugated linoleic acid. Free Radic Biol Med 2014; 72:66-75. [PMID: 24721151 PMCID: PMC4108078 DOI: 10.1016/j.freeradbiomed.2014.03.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/11/2014] [Accepted: 03/29/2014] [Indexed: 11/21/2022]
Abstract
Conjugated linoleic acid (cLA) is a commercially available weight-loss supplement that is not currently regulated by the U.S. FDA. Numerous studies suggest that cLA mediates protection against diseases including cancer, diabetes, atherosclerosis, immune function, and obesity. Based upon these reports, it was hypothesized that supplementation with cLA would improve heart function in aged wild-type (WT) mice. At 10 months of age, mice were treated with cLA, nitrite, or the combination of the two. Echocardiograms revealed that cardiac function was decreased in aged compared to young WT mice, as determined by percentage of fractional shortening. Also, contrary to the hypothesis, mice that received cLA (6-week treatment) had significantly worse cardiac function compared to controls. This effect was attenuated when mice were cotreated with cLA and nitrite. Taken together, these results suggest that cLA-mediated cardiac injury can be circumvented by nitrite supplementation in a murine model of aging.
Collapse
Affiliation(s)
- Kellianne M Piell
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Natia Qipshidze Kelm
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Megan P Caroway
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Masarath Aman
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Marsha P Cole
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|