1
|
Li D, Ma Q. Ubiquitin-specific protease: an emerging key player in cardiomyopathy. Cell Commun Signal 2025; 23:143. [PMID: 40102846 PMCID: PMC11921692 DOI: 10.1186/s12964-025-02123-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Protein quality control (PQC) plays a vital role in maintaining normal heart function, as cardiomyocytes are relatively sensitive to misfolded or damaged proteins, which tend to accumulate under pathological conditions. Ubiquitin-specific protease (USP) is the largest deubiquitinating enzyme family and a key component of the ubiquitin proteasome system (UPS), which is a non-lysosomal protein degradation machinery to mediate PQC in cells. USPs regulate the stability or activity of the target proteins that involve intracellular signaling, transcriptional control of inflammation, antioxidation, and cell growth. Recent studies demonstrate that the USPs can regulate fibrosis, lipid metabolism, glucose homeostasis, hypertrophic response, post-ischemic recovery and cell death such as apoptosis and ferroptosis in cardiomyocytes. Since myocardial cell loss is an important component of cardiomyopathy, therefore, these findings suggest that the UPSs play emerging roles in cardiomyopathy. This review briefly summarizes recent literature on the regulatory roles of USPs in the occurrence and development of cardiomyopathy, giving us new insights into the molecular mechanisms of USPs in different cardiomyopathy and potential preventive strategies for cardiomyopathy.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Qilin Ma
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China.
| |
Collapse
|
2
|
Hu F, Cheng Y, Fan B, Li W, Ye B, Wu Z, Tan Z, He Z. Ruminal microbial metagenomes and host transcriptomes shed light on individual variability in the growth rate of lambs before weaning: the regulated mechanism and potential long-term effect on the host. mSystems 2024; 9:e0087324. [PMID: 39162524 PMCID: PMC11406974 DOI: 10.1128/msystems.00873-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/14/2024] [Indexed: 08/21/2024] Open
Abstract
Weaning weight is a reflection of management during the breastfeeding phase and will influence animal performance in subsequent phases, considered important indicators within production systems. The aims of this study were as follows: (i) to investigate variability in the growth rate among individual lambs from ewes rearing single or twin lambs fed with two different diets and (ii) to explore the molecular mechanisms regulating the growth rate and the potential long-term effects on the host. No significant change in lamb average daily gain (ADG) was observed in litter size and diet treatment, and there were large variations among individual lambs (ranging from 0.13 to 0.41 kg/day). Further analysis was conducted on serum amino acids, rumen fermentation characteristics, rumen metagenomics and transcriptome, and hepatic transcriptome of lambs with extremely high (HA; n = 6) and low (LA; n = 6) ADG. We observed significant increases in serum lysine, leucine, alanine, and phenylalanine in the HA group. The metagenome revealed that the HA group presented a higher rumen propionate molar proportion via increasing gene abundance in the succinate pathway for propionate synthesis. For the rumen transcriptome, higher expressed gene sets in the HA group were mainly related to rumen epithelial growth, including cytokine-cytokine receptor interaction, Jak-STAT signaling pathway, and adherens junction. For the liver transcriptome, the upregulated KEGG pathways in the HA group were primarily associated with fatty acid degradation, glyoxylate and dicarboxylate metabolism, cholesterol metabolism, and the immune system. This research suggests that preweaning lambs with high ADG may benefit from rumen development and enhanced liver metabolic and immune function. IMPORTANCE There is accumulating evidence indicating that the early-life rumen microbiome plays vital roles in rumen development and microbial fermentation, which subsequently affects the growth of young ruminants. The liver is also vital to regulate the metabolism and distribution of nutrients. Our results demonstrate that lambs with high average daily gain (ADG) enhanced microbial volatile fatty acid (VFA) metabolism toward rumen propionate and serum amino acid (AA) production to support host growth. The study highlights that high ADG in the preweaning period is beneficial for the rumen development and liver energy metabolism, leading to better growth later in life. Overall, this study explores the molecular mechanisms regulating the growth rate and the potential long-term effects of increased growth rate on the host metabolism, providing fundamental knowledge about nutrient manipulation in pre-weaning.
Collapse
Affiliation(s)
- Fan Hu
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan Cheng
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bing Fan
- Hulun Buir State Farm Technology Development, Hailar, China
| | - Wei Li
- Hulun Buir State Farm Tenihe Farm, Hulun Buir, China
| | - Bingsen Ye
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
| | - Zhiwu Wu
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiliang Tan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhixiong He
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Li L, Ma SR, Yu ZL. Targeting the lipid metabolic reprogramming of tumor-associated macrophages: A novel insight into cancer immunotherapy. Cell Oncol (Dordr) 2024; 47:415-428. [PMID: 37776422 DOI: 10.1007/s13402-023-00881-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Tumor-associated macrophages, as the major immunocytes in solid tumors, show divided loyalty and remarkable plasticity in tumorigenesis. Once the M2-to-M1 repolarization is achieved, they could be switched from the supporters for tumor development into the guardians for host immunity. Meanwhile, Lipid metabolic reprogramming is demonstrated to be one of the most important hallmarks of tumor-associated macrophages, which plays a decisive role in regulating their phenotypes and functions to promote tumorigenesis and immunotherapy resistance. Therefore, targeting the lipid metabolism of TAMs may provide a new direction for anti-tumor strategies. CONCLUSION In this review, we first summarized the origins, classifications and general lipid metabolic process of TAMs. Then we discussed the currently available drugs and interventions that target lipid metabolic disorders of TAMs, including those targeting lipid uptake, efflux, lipolysis, FAO and lipid peroxidation. Besides, based on the recent research status, we summarized the present challenges for this cancer immunotherapy, including the precise drug delivery system, the lipid metabolic heterogeneity, and the intricate lipid metabolic interactions in the TME, and we also proposed corresponding possible solutions. Collectively, we hope this review will give researchers a better understanding of the lipid metabolism of TAMs and lead to the development of corresponding anti-tumor therapies in the future.
Collapse
Affiliation(s)
- Liang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, People's Republic of China
| | - Si-Rui Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
4
|
Singh V. F 1F o adenosine triphosphate (ATP) synthase is a potential drug target in non-communicable diseases. Mol Biol Rep 2023; 50:3849-3862. [PMID: 36715790 DOI: 10.1007/s11033-023-08299-3] [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: 11/26/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023]
Abstract
F1Fo adenosine triphosphate (ATP) synthase, also known as the complex V, is the central ATP-producing unit in the cells arranged in the mitochondrial and plasma membranes. F1Fo ATP synthase also regulates the central metabolic processes in the human body driven by proton motive force (Δp). Numerous studies have immensely contributed toward highlighting its regulation in improving energy homeostasis and maintaining mitochondrial integrity, which otherwise gets compromised in illnesses. Yet, its role in the implication of non-communicable diseases remains unknown. F1Fo ATP synthase dysregulation at gene level leads to reduced activity and delocalization in the cristae and plasma membranes, which is directly associated with non-communicable diseases: cardiovascular diseases, diabetes, neurodegenerative disorders, cancer, and renal diseases. Individual subunits of the F1Fo ATP synthase target ligand-based competitive or non-competitive inhibition. After performing a systematic literature review to understand its specific functions and its novel drug targets, the present article focuses on the central role of F1Fo ATP synthase in primary non-communicable diseases. Next, it discusses its involvement through various pathways and the effects of multiple inhibitors, activators, and modulators specific to non-communicable diseases with a futuristic outlook.
Collapse
Affiliation(s)
- Varsha Singh
- Centre for Life Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
5
|
Structural and functional characterization of turbot pparγ: Activation during high temperature and regulation of lipid metabolism. J Therm Biol 2022; 108:103279. [DOI: 10.1016/j.jtherbio.2022.103279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/26/2022] [Accepted: 06/03/2022] [Indexed: 11/18/2022]
|
6
|
Maiuolo J, Carresi C, Gliozzi M, Musolino V, Scarano F, Coppoletta AR, Guarnieri L, Nucera S, Scicchitano M, Bosco F, Ruga S, Zito MC, Macri R, Cardamone A, Serra M, Mollace R, Tavernese A, Mollace V. Effects of Bergamot Polyphenols on Mitochondrial Dysfunction and Sarcoplasmic Reticulum Stress in Diabetic Cardiomyopathy. Nutrients 2021; 13:nu13072476. [PMID: 34371986 PMCID: PMC8308586 DOI: 10.3390/nu13072476] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is the leading cause of death and disability in the Western world. In order to safeguard the structure and the functionality of the myocardium, it is extremely important to adequately support the cardiomyocytes. Two cellular organelles of cardiomyocytes are essential for cell survival and to ensure proper functioning of the myocardium: mitochondria and the sarcoplasmic reticulum. Mitochondria are responsible for the energy metabolism of the myocardium, and regulate the processes that can lead to cell death. The sarcoplasmic reticulum preserves the physiological concentration of the calcium ion, and triggers processes to protect the structural and functional integrity of the proteins. The alterations of these organelles can damage myocardial functioning. A proper nutritional balance regarding the intake of macronutrients and micronutrients leads to a significant improvement in the symptoms and consequences of heart disease. In particular, the Mediterranean diet, characterized by a high consumption of plant-based foods, small quantities of red meat, and high quantities of olive oil, reduces and improves the pathological condition of patients with heart failure. In addition, nutritional support and nutraceutical supplementation in patients who develop heart failure can contribute to the protection of the failing myocardium. Since polyphenols have numerous beneficial properties, including anti-inflammatory and antioxidant properties, this review gathers what is known about the beneficial effects of polyphenol-rich bergamot fruit on the cardiovascular system. In particular, the role of bergamot polyphenols in mitochondrial and sarcoplasmic dysfunctions in diabetic cardiomyopathy is reported.
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Anna Rita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Lorenza Guarnieri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Caterina Zito
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Roberta Macri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Serra
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Rocco Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
| | - Annamaria Tavernese
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-327-475-8006
| |
Collapse
|
7
|
Liu Z, Ding J, McMillen TS, Villet O, Tian R, Shao D. Enhancing fatty acid oxidation negatively regulates PPARs signaling in the heart. J Mol Cell Cardiol 2020; 146:1-11. [PMID: 32592696 DOI: 10.1016/j.yjmcc.2020.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022]
Abstract
High fatty acid oxidation (FAO) is associated with lipotoxicity, but whether it causes lipotoxic cardiomyopathy remains controversial. Molecular mechanisms that may be responsible for FAO-induced lipotoxic cardiomyopathy are also elusive. In this study, increasing FAO by genetic deletion of acetyl-CoA carboxylase 2 (ACC2) did not induce cardiac dysfunction after 16 weeks of high fat diet (HFD) feeding. This suggests that increasing FAO, per se, does not cause metabolic cardiomyopathy in obese mice. We compared transcriptomes of control and ACC2 deficient mouse hearts under chow- or HFD-fed conditions. ACC2 deletion had a significant impact on the global transcriptome including downregulation of the peroxisome proliferator-activated receptors (PPARs) signaling and fatty acid degradation pathways. Increasing fatty acids by HFD feeding normalized expression of fatty acid degradation genes in ACC2 deficient mouse hearts to the same level as the control mice. In contrast, cardiac transcriptome analysis of the lipotoxic mouse model (db/db) showed an upregulation of PPARs signaling and fatty acid degradation pathways. Our results suggest that enhancing FAO by genetic deletion of ACC2 negatively regulates PPARs signaling through depleting endogenous PPAR ligands, which can serve as a negative feedback mechanism to prevent excess activation of PPAR signaling under non-obese condition. In obesity, excessive lipid availability negates the feedback mechanism resulting in over activation of PPAR cascade, thus contributes to the development of cardiac lipotoxicity.
Collapse
Affiliation(s)
- ZhengLong Liu
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA 98109, USA
| | - Jeffrey Ding
- Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA 92093, USA
| | - Timothy S McMillen
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA 98109, USA
| | - Outi Villet
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA 98109, USA
| | - Rong Tian
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA 98109, USA.
| | - Dan Shao
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
8
|
Yi S, Chen P, Yang L, Zhu L. Probing the hepatotoxicity mechanisms of novel chlorinated polyfluoroalkyl sulfonates to zebrafish larvae: Implication of structural specificity. ENVIRONMENT INTERNATIONAL 2019; 133:105262. [PMID: 31665679 DOI: 10.1016/j.envint.2019.105262] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 06/10/2023]
Abstract
Hepatotoxicity in zebrafish (Danio rerio) larvae elicited by legacy perfluorooctane sulfonate (PFOS) and its three novel chlorinated alternatives, including chlorinated polyfluorooctane sulfonate (Cl-PFOS) and chlorinated polyfluoroalkyl ether sulfonates (6:2 and 8:2 Cl-PFESA analogs), was evaluated in this study. Upon 7-day separate exposure to the four target compounds at 1 µmol/L, significant hepatic steatosis in exposed larvae was evidenced by pathological micro/macro vacuolation, which was presumably attributed to the excess accumulation of lipid, especially the overloaded triglyceride (TG) level. Disruption on gene transcription was subjected to a structure-dependent manner. In general, PFOS, Cl-PFOS and 6:2 Cl-PFESA of the identical carbon chain length (i.e. C8), despite with different substituents, displayed a similar activation mode and comparable disruptive potency on lipid metabolism responsive genes, which particularly promoted fatty acid synthesis (acetyl-CoA carboxylase, acacb) and β-oxidation (cytochrome P450 enzymes-1A, cyp1a; peroxisomal acyl-CoA oxidase 1, acox1; and acyl-CoA dehy-drogenase, acadm). However, 8:2 Cl-PFESA with a prolonged carbon chain length (i.e. C10), preferentially disturbed fatty acid exportation (apolipoprotein-B100, apob) and triggered a different modulation pattern on fatty acid β-oxidation against the other three compounds. Molecular docking analysis indicated that 8:2 Cl-PFESA exhibited considerably higher peroxisome proliferator-activated receptors (PPARs) antagonism than others, corresponding to its unique suppression effect on fatty acid β-oxidation responsive genes. To our knowledge, this is the first in vivo study reporting hepatotoxicity of Cl-PFOS and Cl-PFESAs to aquatic organisms. Although characterized with different toxic mode-of-action, these novel alternatives can elicit hepatic steatosis as strong as PFOS, stressing the biological risks in view of their global contamination.
Collapse
Affiliation(s)
- Shujun Yi
- State Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Pengyu Chen
- State Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Liping Yang
- State Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Lingyan Zhu
- State Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| |
Collapse
|
9
|
Li L, Jing L, Zhao J, Lv J, Yang W, Li W, Zhou L. Valsartan inhibits RhoA-ROCK2-MYL pathway in rat model of alcoholic cardiomyopathy. Exp Ther Med 2019; 18:4313-4321. [PMID: 31777538 PMCID: PMC6862588 DOI: 10.3892/etm.2019.8079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/01/2018] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate variations in the Ras homolog gene family, member A (RhoA)-Rho-associated protein kinase 2 (ROCK2)-myosin light chain (MYL) pathway in a rat model of alcoholic cardiomyopathy (ACM) and the role of angiotensin-converting enzyme inhibitor drugs. Rat models of ACM were established via alcoholic gavage + free access to alcohol. The structural and functional changes of the heart were analyzed by hematoxylin-eosin staining, Masson's trichrome staining, immunohistochemistry staining, western blotting and fluorescence quantitative polymerase chain reaction. A total of 16 weeks later, a decreased ejection fraction and left ventricular fractional shortening in the alcohol group compared with the control group were demonstrated resulting in an increased left ventricular end diastolic diameter. These adverse effects were ameliorated following treatment with valsartan. In addition, the alcohol group revealed a disorganized arrangement of myocardial filaments, which was improved upon treatment with valsartan. RhoA and ROCK2 protein expression significantly increased in myocardial cells in the alcohol compared with the control group. Following drug intervention with valsartan, expression of RhoA and ROCK2 proteins were inhibited in the alcohol group. Furthermore, significantly elevated RhoA and ROCK2 and decreased MYL protein and mRNA expression in the alcohol group was demonstrated compared with the control group. Administration of valsartan reversed the expression profile of RhoA, ROCK and MYL in ACM. Expression of RhoA and ROCK were elevated with downregulation of MYL resulting in heart failure. However, the angiotensin receptor antagonist diminished the expression of RhoA and ROCK and enhanced the expression of MYL. The results of the present study suggest a curative effect of valsartan in ACM.
Collapse
Affiliation(s)
- Luyifei Li
- Department of Internal Critical Illness, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Ling Jing
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Jiyi Zhao
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Jiachen Lv
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Wen Yang
- Department of The First Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Weimin Li
- Department of The Fifth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Lijun Zhou
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| |
Collapse
|
10
|
Lee TI, Kao YH, Baigalmaa L, Lee TW, Lu YY, Chen YC, Chao TF, Chen YJ. Sodium hydrosulphide restores tumour necrosis factor-α-induced mitochondrial dysfunction and metabolic dysregulation in HL-1 cells. J Cell Mol Med 2019; 23:7641-7650. [PMID: 31496037 PMCID: PMC6815823 DOI: 10.1111/jcmm.14637] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/27/2019] [Accepted: 08/06/2019] [Indexed: 01/22/2023] Open
Abstract
Tumour necrosis factor (TNF)‐α induces cardiac metabolic disorder and mitochondrial dysfunction. Hydrogen sulphide (H2S) contains anti‐inflammatory and biological effects in cardiomyocytes. This study investigated whether H2S modulates TNF‐α‐dysregulated mitochondrial function and metabolism in cardiomyocytes. HL‐1 cells were incubated with TNF‐α (25 ng/mL) with or without sodium hydrosulphide (NaHS, 0.1 mmol/L) for 24 hours. Cardiac peroxisome proliferator‐activated receptor (PPAR) isoforms, pro‐inflammatory cytokines, receptor for advanced glycation end products (RAGE) and fatty acid metabolism were evaluated through Western blotting. The mitochondrial oxygen consumption rate and adenosine triphosphate (ATP) production were investigated using Seahorse XF24 extracellular flux analyzer and bioluminescence assay. Fluorescence intensity using 2′, 7′‐dichlorodihydrofluorescein diacetate was used to evaluate mitochondrial oxidative stress. NaHS attenuated the impaired basal and maximal respiration, ATP production and ATP synthesis and enhanced mitochondrial oxidative stress in TNF‐α‐treated HL‐1 cells. TNF‐α‐treated HL‐1 cells exhibited lower expression of PPAR‐α, PPAR‐δ, phosphorylated 5′ adenosine monophosphate‐activated protein kinase‐α2, phosphorylated acetyl CoA carboxylase, carnitine palmitoyltransferase‐1, PPAR‐γ coactivator 1‐α and diacylglycerol acyltransferase 1 protein, but higher expression of PPAR‐γ, interleukin‐6 and RAGE protein than control or combined NaHS and TNF‐α‐treated HL‐1 cells. NaHS modulates the effects of TNF‐α on mitochondria and the cardiometabolic system, suggesting its therapeutic potential for inflammation‐induced cardiac dysfunction.
Collapse
Affiliation(s)
- Ting-I Lee
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Lkhagva Baigalmaa
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Tze-Fan Chao
- Department of Medicine, Heart Rhythm Center and Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical Univsersity, Taipei, Taiwan
| |
Collapse
|
11
|
Cardiomyocyte mitochondrial dysfunction in diabetes and its contribution in cardiac arrhythmogenesis. Mitochondrion 2019; 46:6-14. [DOI: 10.1016/j.mito.2019.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/16/2019] [Accepted: 03/20/2019] [Indexed: 01/09/2023]
|
12
|
Zhang H, He J, Li N, Gao N, Du Q, Chen B, Chen F, Shan X, Ding Y, Zhu W, Wu Y, Tang J, Jia X. Lipid accumulation responses in the liver of Rana nigromaculata induced by perfluorooctanoic acid (PFOA). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 167:29-35. [PMID: 30292973 DOI: 10.1016/j.ecoenv.2018.09.120] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 05/22/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a perfluorinated compound that is widely distributed, is persistent in the environment, and has a low-level chronic exposure effect on human health. The aim of this study was to investigate the peroxisome proliferator activated receptors γ (PPARγ) and the sterol regulatory element-binding protein 2 (SREBP2) signaling pathways in regulating the lipid damage response to PFOA in the livers of amphibians. Male and female frogs (Rana nigromaculata) were exposed to 0, 0.01, 0.1, 0.5 and 1 mg/L PFOA. After treatment, we evaluated the pathological changes in the liver by Oil Red O, staining and examined the total cholesterol (T-CHO) and triglyceride (TG) contents. The mRNA expression levels of PPARγ, Fatty acid synthase (FAS), Acetyl-CoA carboxylase (ACC), Glycerol-3-phosphate acyltransferase (GPAT), SREBP2 and 3-hydroxy-3-methylglutaryl CoA (HMG-CoA) were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The administration of PFOA caused marked lipid accumulation damage in the amphibian livers. The T-CHO contents were elevated significantly after PFOA treatment; these results show a dose-dependent manner in both sexes. The TG content showed a significant increase in male livers, while it was elevated significantly in female livers. The RT-PCR results showed that the mRNA expression levels of PPARγ, ACC, FAS, GPAT, SREBP2 and HMG-CoA were significantly dose-dependently increased in the PFOA-treated groups compared with those of the control group. Our results demonstrated that PFOA-induced lipid accumulation also affected the expression levels of genes FAS, ACC, GPAT and HMG-CoA in the PPARγ and SREBP2 signaling pathways in the liver. These finding will provide a scientific theoretical basis for the protection of Rana nigromaculata against PFOA effects.
Collapse
Affiliation(s)
- Hangjun Zhang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China; Guangzhou Key Laboratory of Environmental Exposure and Health, School of Environment, Jinan University, Guangzhou 510632, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou, Zhejiang Province 310036, China
| | - Jianbo He
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Ning Li
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Nana Gao
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Qiongxia Du
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Bin Chen
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Feifei Chen
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Xiaodong Shan
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Ying Ding
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou, Zhejiang Province 310036, China
| | - Weiqin Zhu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou, Zhejiang Province 310036, China
| | - Yingzhu Wu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Juan Tang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China
| | - Xiuying Jia
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province 310036, China.
| |
Collapse
|
13
|
Abstract
The nuclear receptor peroxisome proliferator-activated receptor δ (PPARδ) can transcriptionally regulate target genes. PPARδ exerts essential regulatory functions in the heart, which requires constant energy supply. PPARδ plays a key role in energy metabolism, controlling not only fatty acid (FA) and glucose oxidation, but also redox homeostasis, mitochondrial biogenesis, inflammation, and cardiomyocyte proliferation. PPARδ signaling is impaired in the heart under various pathological conditions, such as pathological cardiac hypertrophy, myocardial ischemia/reperfusion, doxorubicin cardiotoxicity and diabetic cardiomyopathy. PPARδ deficiency in the heart leads to cardiac dysfunction, myocardial lipid accumulation, cardiac hypertrophy/remodeling and heart failure. This article provides an up-today overview of this research area and discusses the role of PPARδ in the heart in light of the complex mechanisms of its transcriptional regulation and its potential as a translatable therapeutic target for the treatment of cardiac disorders.
Collapse
Affiliation(s)
- Qinglin Yang
- Cardiovascular Center of Excellence, LSU Healther Science Center, 533 Bolivar St, New Orleans, LA 70112, USA
| | - Qinqiang Long
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| |
Collapse
|
14
|
Xu Y, Zhang Y, Ye J. IL-6: A Potential Role in Cardiac Metabolic Homeostasis. Int J Mol Sci 2018; 19:ijms19092474. [PMID: 30134607 PMCID: PMC6164544 DOI: 10.3390/ijms19092474] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022] Open
Abstract
Interleukin-6 (IL-6) is implicated in multiple biological functions including immunity, neural development, and haematopoiesis. Recently, mounting evidence indicates that IL-6 plays a key role in metabolism, especially lipid metabolic homeostasis. A working heart requires a high and constant energy input which is largely generated by fatty acid (FA) β-oxidation. Under pathological conditions, the precise balance between cardiac FA uptake and metabolism is perturbed so that excessive FA is accumulated, thereby predisposing to myocardial dysfunction (cardiac lipotoxicity). In this review, we summarize the current evidence that suggests the involvement of IL-6 in lipid metabolism. Cardiac metabolic features and consequences of myocardial lipotoxicity are also briefly analyzed. Finally, the roles of IL-6 in cardiac FA uptake (i.e., serum lipid profile and myocardial FA transporters) and FA metabolism (namely, β-oxidation, mitochondrial function, biogenesis, and FA de novo synthesis) are discussed. Overall, understanding how IL-6 transmits signals to affect lipid metabolism in the heart might allow for development of better clinical therapies for obesity-associated cardiac lipotoxicity.
Collapse
Affiliation(s)
- Yitao Xu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London W120NN, UK.
| | - Yubin Zhang
- State Key laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| | - Junmei Ye
- State Key laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| |
Collapse
|
15
|
Sikder K, Shukla SK, Patel N, Singh H, Rafiq K. High Fat Diet Upregulates Fatty Acid Oxidation and Ketogenesis via Intervention of PPAR-γ. Cell Physiol Biochem 2018; 48:1317-1331. [PMID: 30048968 PMCID: PMC6179152 DOI: 10.1159/000492091] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/25/2018] [Indexed: 01/07/2023] Open
Abstract
Background/Aims: Systemic hyperlipidemia and intracellular lipid accumulation induced by chronic high fat diet (HFD) leads to enhanced fatty acid oxidation (FAO) and ketogenesis. The present study was aimed to determine whether activation of peroxisome proliferator-activated receptor-γ (PPAR-γ) by surplus free fatty acids (FA) in hyperlipidemic condition, has a positive feedback regulation over FAO and ketogenic enzymes controlling lipotoxicity and cardiac apoptosis. Methods: 8 weeks old C57BL/6 wild type (WT) or PPAR-γ−/− mice were challenged with 16 weeks 60% HFD to induce obesity mediated type 2 diabetes mellitus (T2DM) and diabetic cardiomyopathy. Treatment course was followed by echocardiographic measurements, glycemic and lipid profiling, immunoblot, qPCR and immunohistochemistry (IHC) analysis of PPAR-γ and following mitochondrial metabolic enzymes 3-hydroxy-3- methylglutaryl-CoA synthase (HMGCS2), mitochondrial β-hydroxy butyrate dehydrogenase (BDH1) and pyruvate dehydrogenase kinase isoform 4 (PDK4). In vivo model was translated in vitro, with neonatal rat cardiomyocytes (NRCM) treated with PPAR-γ agonist/antagonist and PPAR-γ overexpression adenovirus in presence of palmitic acid (PA). Apoptosis was determined in vivo from left ventricular heart by TUNEL assay and immunoblot analysis. Results: We found exaggerated circulating ketone bodies production and expressions of the related mitochondrial enzymes HMGCS2, BDH1 and PDK4 in HFD-induced diabetic hearts and in PA-treated NRCM. As a mechanistic approach we found HFD mediated activation of PPAR-03B3 is associated with the above-mentioned mitochondrial enzymes. HFD-fed PPAR-γ−/− mice display decreased hyperglycemia, hyperlipidemia associated with increased insulin responsiveness as compared to HFD-fed WT mice PPAR-γ−/−−HFD mice demonstrated a more robust functional recovery after diabetes induction, as well as significantly reduced myocyte apoptosis and improved cardiac function. Conclusions: PPAR-γ has been described previously to regulate lipid metabolism and adipogenesis. The present study suggests for the first time that increased PPAR-γ expression by HFD is responsible for cardiac dysfunction via upregulation of mitochondrial enzymes HMGCS2, BDH1 and PDK4. Targeting PPAR-γ and its downstream mitochondrial enzymes will provide novel strategies in preventing metabolic and myocardial dysfunction in diabetes mellitus.
Collapse
Affiliation(s)
- Kunal Sikder
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sanket Kumar Shukla
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Neel Patel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Khadija Rafiq
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang T, Wang X. Liraglutide protects cardiac function in diabetic rats through the PPARα pathway. Biosci Rep 2018; 38:BSR20180059. [PMID: 29440457 PMCID: PMC5857913 DOI: 10.1042/bsr20180059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 01/28/2018] [Accepted: 02/09/2018] [Indexed: 01/08/2023] Open
Abstract
Increasing evidence shows that diabetes causes cardiac dysfunction. We hypothesized that a glucagon-like peptide-1 analogue, liraglutide, would attenuate cardiac dysfunction in diabetic rats. Twenty-four Sprague Dawley (SD) rats were divided into 2 groups fed either a normal diet (normal, n = 6) or a high-fat diet (HFD, n = 18) for 4 weeks. Then, the HFD rats were injected with streptozotocin (STZ) to create a diabetic rat model. Diabetic rats were divided into 3 subgroups receiving vehicle (diabetic, n = 6), a low dose of liraglutide (Llirag, 0.2 mg/kg/day, n = 6) or a high dose of liraglutide (Hlirag, 0.4 mg/kg/day, n = 6). Metabolic parameters, systolic blood pressure, heart rate, left ventricular (LV) function, and whole genome expression of the heart were determined. Diabetic rats developed insulin resistance, increased blood lipid levels and oxidative stress, and impaired LV function, serum adiponectin, NO. Liraglutide improved insulin resistance, serum adiponectin, NO, heart rate and LV function and reduced blood triglyceride, total cholesterol levels and oxidative stress. Moreover, liraglutide increased heart Nr1h3 , Ppar-α and Srebp expression and reduced Dgat , and Angptl3 expression. Liraglutide prevented in cardiac dysfunction by activating the PPARα pathway to inhibit Dgat expression and oxidative stress in diabetic rats.
Collapse
Affiliation(s)
- Qian Zhang
- Peking Union Medical College Hospital, Beijing, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jia Zheng
- Peking Union Medical College Hospital, Beijing, China
| | - Ming Li
- Peking Union Medical College Hospital, Beijing, China
| | - Miao Yu
- Peking Union Medical College Hospital, Beijing, China
| | - Fan Ping
- Peking Union Medical College Hospital, Beijing, China
| | - Tong Wang
- Peking Union Medical College Hospital, Beijing, China
| | - Xiaojing Wang
- Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
17
|
Maccallini C, Mollica A, Amoroso R. The Positive Regulation of eNOS Signaling by PPAR Agonists in Cardiovascular Diseases. Am J Cardiovasc Drugs 2017; 17:273-281. [PMID: 28315197 DOI: 10.1007/s40256-017-0220-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Increasing evidence shows that activation of peroxisome proliferator-activated receptors (PPARs) plays an essential role in the regulation of vascular endothelial function through a range of mechanisms, including non-metabolic. Among these, the PPAR-mediated activation of endothelial nitric oxide synthase (eNOS) appears to be of considerable importance. The regulated and sustained bioavailability of nitric oxide (NO) in the endothelium is essential to avoid the development of cardiovascular diseases such as hypertension or atherosclerosis. Therefore, a deeper understanding of the different effects of specific PPAR ligands on NO bioavailability could be useful in the development of novel or multi-targeted PPAR agonists. In this review, we report the most meaningful and up-to-date in vitro and in vivo studies of the regulation of NO production performed by different PPAR agonists. Insights into the molecular mechanisms of PPAR-mediated eNOS activation are also provided. Although findings from animal studies in which the activation of PPARα, PPARβ/δ, or PPARγ have provided clear vasoprotective effects have been promising, several benefits from PPAR agonists are offset by unwanted outcomes. Therefore, new insights could be useful in the development of tissue-targeted PPAR agonists with more tolerable side effects to improve treatment options for cardiovascular diseases.
Collapse
|
18
|
Yan P, Chen K, Wang Q, Yang D, Li D, Yang Y. UCP-2 is involved in angiotensin-II-induced abdominal aortic aneurysm in apolipoprotein E-knockout mice. PLoS One 2017; 12:e0179743. [PMID: 28683125 PMCID: PMC5500278 DOI: 10.1371/journal.pone.0179743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 06/02/2017] [Indexed: 02/07/2023] Open
Abstract
UCP-2 shows an important role in modulating of mitochondrial membrane potential and cell apoptosis. Whether or not UCP-2 could been a critical factor in preventing AAA formation is not known. We report that UCP-2 protein and mRNA expression were significantly higher in Ang-Ⅱ-induced AAA of mice. The incident rate of AAA in UCP-2-/-ApoE-/- mice after Ang-Ⅱtreatment was higher than the rate in the UCP-2+/+ApoE-/- mice. The abdominal aorta from UCP-2-/-ApoE-/- mice showed the medial hypertrophy, fragmentation of elastic lamellas and depletion of α-SMA. The NADPH oxidase activity and level of MDA was significantly higher in UCP-2-/-ApoE-/- mice than UCP-2+/+ApoE-/- or WT mice. Besides, the SOD activity is increased in UCP-2+/+ApoE-/- mice as compared with WT mice, whereas deficiency of UCP-2 decreased the increasing SOD activity in Ang-Ⅱ treated ApoE-/- mice. UCP-2 knockout up-regulated the MMP2 and MMP9 expression in aortic aneurysm. Ang-Ⅱ induced apoptosis of VSMCs was increased in UCP-2-/-ApoE-/- mice. And the expression of eNOS in vascular tissue from UCP-2-/-ApoE-/- mice is lower than WT and UCP-2+/+ApoE-/- mice. This study provides a mechanism by which UCP-2, via anti-oxidants and anti-apoptosis, participates in the preventing of AAA formation.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Angiotensin II/pharmacology
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Apoptosis/drug effects
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation
- Malondialdehyde/metabolism
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Membrane Potential, Mitochondrial/drug effects
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- NADPH Oxidases/genetics
- NADPH Oxidases/metabolism
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Superoxide Dismutase/genetics
- Superoxide Dismutase/metabolism
- Uncoupling Protein 2/deficiency
- Uncoupling Protein 2/genetics
Collapse
Affiliation(s)
- Peng Yan
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Ken Chen
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Qiang Wang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Dachun Yang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - De Li
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Yongjian Yang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
- * E-mail:
| |
Collapse
|
19
|
Zhang Y, Liao P, Zhu M, Li W, Hu D, Guan S, Chen L. Baicalin Attenuates Cardiac Dysfunction and Myocardial Remodeling in a Chronic Pressure-Overload Mice Model. Cell Physiol Biochem 2017; 41:849-864. [DOI: 10.1159/000459708] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/27/2016] [Indexed: 11/19/2022] Open
Abstract
Background/Aims: Baicalin has been shown to be effective for various animal models of cardiovascular diseases, such as pulmonary hypertension, atherosclerosis and myocardial ischaemic injury. However, whether baicalin plays a role in cardiac hypertrophy remains unknown. Here we investigated the protective effects of baicalin on cardiac hypertrophy induced by pressure overload and explored the potential mechanisms involved. Methods: C57BL/6J-mice were treated with baicalin or vehicle following transverse aortic constriction or Sham surgery for up to 8 weeks, and at different time points, cardiac function and heart size measurement and histological and biochemical examination were performed. Results: Mice under pressure overload exhibited cardiac dysfunction, high mortality, myocardial hypertrophy, increased apoptosis and fibrosis markers, and suppressed cardiac expression of PPARα and PPARβ/δ. However, oral administration of baicalin improved cardiac dysfunction, decreased mortality, and attenuated histological and biochemical changes described above. These protective effects of baicalin were associated with reduced heart and cardiomyocyte size, lower fetal genes expression, attenuated cardiac fibrosis, lower expression of profibrotic markers, and decreased apoptosis signals in heart tissue. Moreover, we found that baicalin induced PPARα and PPARβ/δ expression in vivo and in vitro. Subsequent experiments demonstrated that long-term baicalin treatment presented no obvious cardiac lipotoxicity. Conclusions: The present results demonstrated that baicalin attenuates pressure overload induced cardiac dysfunction and ventricular remodeling, which would be due to suppressed cardiac hypertrophy, fibrosis, apoptosis and metabolic abnormality.
Collapse
|
20
|
Hathaway QA, Nichols CE, Shepherd DL, Stapleton PA, McLaughlin SL, Stricker JC, Rellick SL, Pinti MV, Abukabda AB, McBride CR, Yi J, Stine SM, Nurkiewicz TR, Hollander JM. Maternal-engineered nanomaterial exposure disrupts progeny cardiac function and bioenergetics. Am J Physiol Heart Circ Physiol 2016; 312:H446-H458. [PMID: 28011589 PMCID: PMC5402018 DOI: 10.1152/ajpheart.00634.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 01/25/2023]
Abstract
Nanomaterial production is expanding as new industrial and consumer applications are introduced. Nevertheless, the impacts of exposure to these compounds are not fully realized. The present study was designed to determine whether gestational nano-sized titanium dioxide exposure impacts cardiac and metabolic function of developing progeny. Pregnant Sprague-Dawley rats were exposed to nano-aerosols (~10 mg/m3, 130- to 150-nm count median aerodynamic diameter) for 7-8 nonconsecutive days, beginning at gestational day 5-6 Physiological and bioenergetic effects on heart function and cardiomyocytes across three time points, fetal (gestational day 20), neonatal (4-10 days), and young adult (6-12 wk), were evaluated. Functional analysis utilizing echocardiography, speckle-tracking based strain, and cardiomyocyte contractility, coupled with mitochondrial energetics, revealed effects of nano-exposure. Maternal exposed progeny demonstrated a decrease in E- and A-wave velocities, with a 15% higher E-to-A ratio than controls. Myocytes isolated from exposed animals exhibited ~30% decrease in total contractility, departure velocity, and area of contraction. Bioenergetic analysis revealed a significant increase in proton leak across all ages, accompanied by decreases in metabolic function, including basal respiration, maximal respiration, and spare capacity. Finally, electron transport chain complex I and IV activities were negatively impacted in the exposed group, which may be linked to a metabolic shift. Molecular data suggest that an increase in fatty acid metabolism, uncoupling, and cellular stress proteins may be associated with functional deficits of the heart. In conclusion, gestational nano-exposure significantly impairs the functional capabilities of the heart through cardiomyocyte impairment, which is associated with mitochondrial dysfunction.NEW & NOTEWORTHY Cardiac function is evaluated, for the first time, in progeny following maternal nanomaterial inhalation. The findings indicate that exposure to nano-sized titanium dioxide (nano-TiO2) during gestation negatively impacts cardiac function and mitochondrial respiration and bioenergetics. We conclude that maternal nano-TiO2 inhalation contributes to adverse cardiovascular health effects, lasting into adulthood.
Collapse
Affiliation(s)
- Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Cody E Nichols
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Danielle L Shepherd
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Phoebe A Stapleton
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Sarah L McLaughlin
- Department of Cancer Cell Biology, West Virginia University School of Medicine; Morgantown, West Virginia; and
| | - Janelle C Stricker
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Stephanie L Rellick
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Mark V Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Alaeddin B Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Carroll R McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Jinghai Yi
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Seth M Stine
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; .,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
21
|
Kaimoto S, Hoshino A, Ariyoshi M, Okawa Y, Tateishi S, Ono K, Uchihashi M, Fukai K, Iwai-Kanai E, Matoba S. Activation of PPAR-α in the early stage of heart failure maintained myocardial function and energetics in pressure-overload heart failure. Am J Physiol Heart Circ Physiol 2016; 312:H305-H313. [PMID: 28011586 DOI: 10.1152/ajpheart.00553.2016] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/07/2016] [Accepted: 11/17/2016] [Indexed: 01/26/2023]
Abstract
Failing heart loses its metabolic flexibility, relying increasingly on glucose as its preferential substrate and decreasing fatty acid oxidation (FAO). Peroxisome proliferator-activated receptor α (PPAR-α) is a key regulator of this substrate shift. However, its role during heart failure is complex and remains unclear. Recent studies reported that heart failure develops in the heart of myosin heavy chain-PPAR-α transgenic mice in a manner similar to that of diabetic cardiomyopathy, whereas cardiac dysfunction is enhanced in PPAR-α knockout mice in response to chronic pressure overload. We created a pressure-overload heart failure model in mice through transverse aortic constriction (TAC) and activated PPAR-α during heart failure using an inducible transgenic model. After 8 wk of TAC, left ventricular (LV) function had decreased with the reduction of PPAR-α expression in wild-type mice. We examined the effect of PPAR-α induction during heart failure using the Tet-Off system. Eight weeks after the TAC operation, LV construction was preserved significantly by PPAR-α induction with an increase in PPAR-α-targeted genes related to fatty acid metabolism. The increase of expression of fibrosis-related genes was significantly attenuated by PPAR-α induction. Metabolic rates measured by isolated heart perfusions showed a reduction in FAO and glucose oxidation in TAC hearts, but the rate of FAO preserved significantly owing to the induction of PPAR-α. Myocardial high-energy phosphates were significantly preserved by PPAR-α induction. These results suggest that PPAR-α activation during pressure-overloaded heart failure improved myocardial function and energetics. Thus activating PPAR-α and modulation of FAO could be a promising therapeutic strategy for heart failure.NEW & NOTEWORTHY The present study demonstrates the role of PPAR-α activation in the early stage of heart failure using an inducible transgenic mouse model. Induction of PPAR-α preserved heart function, and myocardial energetics. Activating PPAR-α and modulation of fatty acid oxidation could be a promising therapeutic strategy for heart failure.
Collapse
Affiliation(s)
- Satoshi Kaimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Makoto Ariyoshi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Yoshifumi Okawa
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Shuhei Tateishi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Kazunori Ono
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Motoki Uchihashi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Kuniyoshi Fukai
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| | - Eri Iwai-Kanai
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and.,Faculty of Health Care, Tenri Health Care University, Nara, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kawaramachi-Hirokoji, Kyoto, Japan; and
| |
Collapse
|
22
|
Liu L, Zhang LI, Lin YE, Bian Y, Gao X, Qu BO, Li Q. 14-3-3γ regulates cell viability and milk fat synthesis in lipopolysaccharide-induced dairy cow mammary epithelial cells. Exp Ther Med 2016; 11:1279-1287. [PMID: 27073437 PMCID: PMC4812431 DOI: 10.3892/etm.2016.3029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/22/2015] [Indexed: 12/27/2022] Open
Abstract
Our previous study demonstrated that 14-3-3γ overexpression was able to inhibit the production of lipopolysaccharide (LPS)-induced cytokines in dairy cow mammary epithelial cells (DCMECs) by inhibiting the activation of nuclear factor-κB (NF-κB) signaling pathways. However, the association between 14-3-3γ overexpression and milk fat synthesis in LPS-induced DCMECs remains unclear. Therefore, the present study investigated the effect of 14-3-3γ on cell viability and milk fat synthesis in LPS-induced DCMECs. The results of the MTT assay and lactate dehydrogenase activity assay demonstrated that 14-3-3γ overexpression was able to attenuate LPS-induced cytotoxicity in DCMECs, and increase the viability of the cells. In addition, the results of reverse transcription-quantitative polymerase chain reaction suggested that mRNA expression levels of genes associated with milk fat synthesis, including sterol regulatory element binding protein (SREBP1), peroxisome proliferator-activated receptor-γ (PPARG), cluster of differentiation 36, acetyl-coA carboxylase (ACC), fatty acid synthase (FAS) and fatty acid binding protein-3, were significantly upregulated in cells overexpressing the 14-3-3γ protein. In addition, as compared with the LPS-treated group, the activities of FAS and ACC were significantly increased. Furthermore, western blotting demonstrated that 14-3-3γ overexpression enhanced the protein expression levels of phosphorylated SREBP1 and PPARG. These results suggested that high levels of 14-3-3γ protein were able to attenuate LPS-induced cell damage and promote milk fat synthesis in LPS-induced DCMECs by increasing the cell viability and upregulating the expression levels of transcription factors associated with milk fat synthesis.
Collapse
Affiliation(s)
- Lixin Liu
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China; College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - L I Zhang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - Y E Lin
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - Yanjie Bian
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - Xuejun Gao
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - B O Qu
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - Qingzhang Li
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| |
Collapse
|
23
|
Barlaka E, Galatou E, Mellidis K, Ravingerova T, Lazou A. Role of Pleiotropic Properties of Peroxisome Proliferator-Activated Receptors in the Heart: Focus on the Nonmetabolic Effects in Cardiac Protection. Cardiovasc Ther 2016; 34:37-48. [DOI: 10.1111/1755-5922.12166] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Eleftheria Barlaka
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Eleftheria Galatou
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Kyriakos Mellidis
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Tanya Ravingerova
- Institute for Heart Research; Slovak Academy of Sciences; Bratislava Slovak Republic
| | - Antigone Lazou
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| |
Collapse
|
24
|
Peroxisome Proliferator-Activated Receptors and the Heart: Lessons from the Past and Future Directions. PPAR Res 2015; 2015:271983. [PMID: 26587015 PMCID: PMC4637490 DOI: 10.1155/2015/271983] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/05/2015] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear family of ligand activated transcriptional factors and comprise three different isoforms, PPAR-α, PPAR-β/δ, and PPAR-γ. The main role of PPARs is to regulate the expression of genes involved in lipid and glucose metabolism. Several studies have demonstrated that PPAR agonists improve dyslipidemia and glucose control in animals, supporting their potential as a promising therapeutic option to treat diabetes and dyslipidemia. However, substantial differences exist in the therapeutic or adverse effects of specific drug candidates, and clinical studies have yielded inconsistent data on their cardioprotective effects. This review summarizes the current knowledge regarding the molecular function of PPARs and the mechanisms of the PPAR regulation by posttranslational modification in the heart. We also describe the results and lessons learned from important clinical trials on PPAR agonists and discuss the potential future directions for this class of drugs.
Collapse
|
25
|
Tepavčević S, Milutinović DV, Macut D, Stojiljković M, Nikolić M, Božić-Antić I, Ćulafić T, Bjekić-Macut J, Matić G, Korićanac G. Cardiac fatty acid uptake and metabolism in the rat model of polycystic ovary syndrome. Endocrine 2015; 50:193-201. [PMID: 25702158 DOI: 10.1007/s12020-015-0558-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/17/2015] [Indexed: 12/20/2022]
Abstract
Polycystic ovary syndrome (PCOS) is associated with an altered plasma lipid profile and increased risk for cardiovascular diseases. We hypothesized that molecular mechanisms underlying cardiac pathology in PCOS involve changes in expression and subcellular localization of several key proteins involved in cardiac lipid transport and metabolism, such as fatty acid transporter CD36, lipin 1, peroxisome proliferator-activated receptor α (PPARα), peroxisome proliferator-activated receptor γ coactivator-1 (PGC1), and carnitine palmitoyltransferase 1 (CPT1). We used the animal model of PCOS obtained by treating female rats with dihydrotestosterone (DHT). Protein levels of CD36, lipin 1, PPARα, PGC1, and antioxidative enzymes were assessed by Western blot in different cardiac cell compartments. Cardiac triglycerides (TG) and lipid peroxidation were also measured. The content of CD36 was decreased in both the cardiac plasma membranes and intracellular pool. On the other hand, total content of cardiac lipin 1 in DHT-treated rats was elevated, in contrast to decreased microsomal lipin 1 content. An increase in nuclear content of lipin 1 was observed together with elevation of nuclear PPARα and PGC1, and an increase in CPT1 expression. However, lipid peroxidation was reduced in the heart, without alterations in antioxidative enzymes expression and cardiac TG content. The results indicate that treatment of female rats with DHT is accompanied by a decrease of fatty acid uptake and a reduction of lipid peroxidation in the heart. The observed elevation of lipin 1, PPARα, PGC1, and CPT1 expression suggests that cardiac fatty acid metabolism is shifted toward mitochondrial beta oxidation.
Collapse
Affiliation(s)
- Snežana Tepavčević
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, PO Box 522, 11001, Belgrade, Republic of Serbia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
He J, Quintana MT, Sullivan J, L Parry T, J Grevengoed T, Schisler JC, Hill JA, Yates CC, Mapanga RF, Essop MF, Stansfield WE, Bain JR, Newgard CB, Muehlbauer MJ, Han Y, Clarke BA, Willis MS. MuRF2 regulates PPARγ1 activity to protect against diabetic cardiomyopathy and enhance weight gain induced by a high fat diet. Cardiovasc Diabetol 2015; 14:97. [PMID: 26242235 PMCID: PMC4526192 DOI: 10.1186/s12933-015-0252-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/30/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In diabetes mellitus the morbidity and mortality of cardiovascular disease is increased and represents an important independent mechanism by which heart disease is exacerbated. The pathogenesis of diabetic cardiomyopathy involves the enhanced activation of PPAR transcription factors, including PPARα, and to a lesser degree PPARβ and PPARγ1. How these transcription factors are regulated in the heart is largely unknown. Recent studies have described post-translational ubiquitination of PPARs as ways in which PPAR activity is inhibited in cancer. However, specific mechanisms in the heart have not previously been described. Recent studies have implicated the muscle-specific ubiquitin ligase muscle ring finger-2 (MuRF2) in inhibiting the nuclear transcription factor SRF. Initial studies of MuRF2-/- hearts revealed enhanced PPAR activity, leading to the hypothesis that MuRF2 regulates PPAR activity by post-translational ubiquitination. METHODS MuRF2-/- mice were challenged with a 26-week 60% fat diet designed to simulate obesity-mediated insulin resistance and diabetic cardiomyopathy. Mice were followed by conscious echocardiography, blood glucose, tissue triglyceride, glycogen levels, immunoblot analysis of intracellular signaling, heart and skeletal muscle morphometrics, and PPARα, PPARβ, and PPARγ1-regulated mRNA expression. RESULTS MuRF2 protein levels increase ~20% during the development of diabetic cardiomyopathy induced by high fat diet. Compared to littermate wildtype hearts, MuRF2-/- hearts exhibit an exaggerated diabetic cardiomyopathy, characterized by an early onset systolic dysfunction, larger left ventricular mass, and higher heart weight. MuRF2-/- hearts had significantly increased PPARα- and PPARγ1-regulated gene expression by RT-qPCR, consistent with MuRF2's regulation of these transcription factors in vivo. Mechanistically, MuRF2 mono-ubiquitinated PPARα and PPARγ1 in vitro, consistent with its non-degradatory role in diabetic cardiomyopathy. However, increasing MuRF2:PPARγ1 (>5:1) beyond physiological levels drove poly-ubiquitin-mediated degradation of PPARγ1 in vitro, indicating large MuRF2 increases may lead to PPAR degradation if found in other disease states. CONCLUSIONS Mutations in MuRF2 have been described to contribute to the severity of familial hypertrophic cardiomyopathy. The present study suggests that the lack of MuRF2, as found in these patients, can result in an exaggerated diabetic cardiomyopathy. These studies also identify MuRF2 as the first ubiquitin ligase to regulate cardiac PPARα and PPARγ1 activities in vivo via post-translational modification without degradation.
Collapse
Affiliation(s)
- Jun He
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA. .,General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China.
| | - Megan T Quintana
- Department of Surgery, University of North Carolina, Chapel Hill, NC, USA.
| | - Jenyth Sullivan
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA.
| | - Traci L Parry
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA.
| | - Trisha J Grevengoed
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA.
| | - Jonathan C Schisler
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA. .,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA.
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Cecelia C Yates
- Department of Health Promotions and Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Rudo F Mapanga
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | - M Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | | | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA. .,Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA. .,Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.
| | - Yipin Han
- East Chapel Hill High School, Chapel Hill, NC, USA.
| | - Brian A Clarke
- Novartis, Novartis Institutes for BioMedical Research, Inc., 400 Technology Square, Boston, MA, 601-4214, USA.
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA. .,McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA.
| |
Collapse
|
27
|
Cao S, Liu Y, Sun W, Zhao L, Zhang L, Liu X, Yu T. Genome-Wide Expression Profiling of Anoxia/Reoxygenation in Rat Cardiomyocytes Uncovers the Role of MitoKATP in Energy Homeostasis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:756576. [PMID: 26171116 PMCID: PMC4485557 DOI: 10.1155/2015/756576] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/08/2014] [Indexed: 12/30/2022]
Abstract
Mitochondrial ATP-sensitive potassium channel (mitoK(ATP)) is a common end effector of many protective stimuli in myocardial ischemia-reperfusion injury (MIRI). However, the specific molecular mechanism underlying its myocardial protective effect is not well elucidated. We characterized an anoxia/reoxygenation (A/R) model using freshly isolated adult rat cardiomyocytes. MitoK(ATP) status was interfered with its specific opener diazoxide (DZ) or blocker 5-hydroxydecanote (5-HD). Digital gene expression (DGE) and bioinformatic analysis were deployed. Three energy metabolism related genes (MT-ND6, Idh2, and Acadl) were upregulated when mitoK(ATP) opened. In addition, as many as 20 differentially expressed genes (DEGs) were significantly enriched in five energy homeostasis correlated pathways (PPAR, TCA cycle, fatty acid metabolism, and peroxisome). These findings indicated that mitoK(ATP) opening in MIRI resulted in energy mobilization, which was confirmed by measuring ATP content in cardiomyocytes. These causal outcomes could be a molecular mechanism of myocardial protection of mitoKATP and suggested that the mitoK(ATP) opening plays a physiologic role in triggering cardiomyocytes' energy homeostasis during MIRI. Strategies of modulating energy expenditure during myocardial ischemia-reperfusion may be promising approaches to reduce MIRI.
Collapse
Affiliation(s)
- Song Cao
- Department of Anesthesiology, Zunyi Medical College, Zunyi 563000, China
- Guizhou Key Laboratory of Anesthesiology and Organ Protection, Zunyi Medical College, Zunyi 563000, China
| | - Yun Liu
- Guizhou Key Laboratory of Anesthesiology and Organ Protection, Zunyi Medical College, Zunyi 563000, China
- Research Center for Medicine & Biology, Zunyi Medical College, Zunyi 563000, China
| | - Wenting Sun
- Department of Anesthesiology, Zunyi Medical College, Zunyi 563000, China
- Guizhou Key Laboratory of Anesthesiology and Organ Protection, Zunyi Medical College, Zunyi 563000, China
| | - Li Zhao
- Department of Anesthesiology, Zunyi Medical College, Zunyi 563000, China
- Guizhou Key Laboratory of Anesthesiology and Organ Protection, Zunyi Medical College, Zunyi 563000, China
| | - Lin Zhang
- Department of Anesthesiology, Zunyi Medical College, Zunyi 563000, China
- Guizhou Key Laboratory of Anesthesiology and Organ Protection, Zunyi Medical College, Zunyi 563000, China
| | - Xinkui Liu
- Department of Anesthesiology, Zunyi Medical College, Zunyi 563000, China
| | - Tian Yu
- Department of Anesthesiology, Zunyi Medical College, Zunyi 563000, China
- Guizhou Key Laboratory of Anesthesiology and Organ Protection, Zunyi Medical College, Zunyi 563000, China
| |
Collapse
|
28
|
Roul D, Recchia FA. Metabolic alterations induce oxidative stress in diabetic and failing hearts: different pathways, same outcome. Antioxid Redox Signal 2015; 22:1502-14. [PMID: 25836025 PMCID: PMC4449624 DOI: 10.1089/ars.2015.6311] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Several authors have proposed a link between altered cardiac energy substrate metabolism and reactive oxygen species (ROS) generation. A cogent evidence of this association has been found in diabetic cardiomyopathy (dCM); however, experimental findings in animal models of heart failure (HF) and in human myocardium also seem to support the coexistence of the two alterations in HF. CRITICAL ISSUES Two important questions remain open: whether pathological changes in metabolism play an important role in enhancing oxidative stress and whether there is a common pathway linking altered substrate utilization and activation of ROS-generating enzymes, independently of the underlying cardiac pathology. In this regard, the comparison between dCM and HF is intriguing, in that these pathological conditions display very different cardiac metabolic phenotypes. RECENT ADVANCES Our literature review on this topic indicates that a vast body of knowledge is now available documenting the relationship between the metabolism of energy substrates and ROS generation in dCM. In some cases, biochemical mechanisms have been identified. On the other hand, only a few and relatively recent studies have explored this phenomenon in HF and their conclusions are not consistent. FUTURE DIRECTIONS Better methods of investigation, especially in vivo, will be necessary to test whether the metabolic fate of certain substrates is causally linked to ROS production. If successful, these studies will place a new emphasis on the potential clinical relevance of metabolic modulators, which might indirectly mitigate cardiac oxidative stress in dCM, HF, and, possibly, in other pathological conditions.
Collapse
Affiliation(s)
- David Roul
- 1Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Fabio A Recchia
- 1Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania.,2Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
29
|
Barlaka E, Görbe A, Gáspár R, Pálóczi J, Ferdinandy P, Lazou A. Activation of PPARβ/δ protects cardiac myocytes from oxidative stress-induced apoptosis by suppressing generation of reactive oxygen/nitrogen species and expression of matrix metalloproteinases. Pharmacol Res 2015; 95-96:102-10. [PMID: 25828396 DOI: 10.1016/j.phrs.2015.03.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 03/15/2015] [Accepted: 03/15/2015] [Indexed: 01/26/2023]
Abstract
Heart failure still remains one of the leading causes of morbidity and mortality worldwide. A major contributing factor is reactive oxygen/nitrogen species (RONS) overproduction which is associated with cardiac remodeling partly through cardiomyocyte apoptosis. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the nuclear receptor superfamily and have been implicated in cardioprotection. However, the molecular mechanisms are largely unexplored. In this study we sought to investigate the potential beneficial effects evoked by activation of PPARβ/δ under the setting of oxidative stress induced by H2O2 in adult rat cardiac myocytes. The selective PPARβ/δ agonist GW0742 inhibited the H2O2-induced apoptosis and increased cell viability. In addition, generation of RONS was attenuated in cardiac myocytes in the presence of PPARβ/δ agonist. These effects were abolished in the presence of the PPARβ/δ antagonist indicating that the effect was through PPARβ/δ receptor activation. Treatment with PPARβ/δ agonist was also associated with attenuation of caspase-3 and PARP cleavage, upregulation of anti-apoptotic Bcl-2 and concomitant downregulation of pro-apoptotic Bax. In addition, activation of PPARβ/δ inhibited the oxidative-stress-induced MMP-2 and MMP-9 mRNA upregulation. It is concluded that PPARβ/δ activation exerts a cytoprotective effect in adult rat cardiac myocytes subjected to oxidative stress via inhibition of oxidative stress, MMP expression, and apoptosis. Our data suggest that the novel connection between PPAR signaling and MMP down-regulation in cardiac myocytes might represent a new target for the management of oxidative stress-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Eleftheria Barlaka
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Greece
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Renáta Gáspár
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - János Pálóczi
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Greece.
| |
Collapse
|
30
|
KLF15 and PPARα Cooperate to Regulate Cardiomyocyte Lipid Gene Expression and Oxidation. PPAR Res 2015; 2015:201625. [PMID: 25815008 PMCID: PMC4357137 DOI: 10.1155/2015/201625] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022] Open
Abstract
The metabolic myocardium is an omnivore and utilizes various carbon substrates to meet its energetic demand. While the adult heart preferentially consumes fatty acids (FAs) over carbohydrates, myocardial fuel plasticity is essential for organismal survival. This metabolic plasticity governing fuel utilization is under robust transcriptional control and studies over the past decade have illuminated members of the nuclear receptor family of factors (e.g., PPARα) as important regulators of myocardial lipid metabolism. However, given the complexity of myocardial metabolism in health and disease, it is likely that other molecular pathways are likely operative and elucidation of such pathways may provide the foundation for novel therapeutic approaches. We previously demonstrated that Kruppel-like factor 15 (KLF15) is an independent regulator of cardiac lipid metabolism thus raising the possibility that KLF15 and PPARα operate in a coordinated fashion to regulate myocardial gene expression requisite for lipid oxidation. In the current study, we show that KLF15 binds to, cooperates with, and is required for the induction of canonical PPARα-mediated gene expression and lipid oxidation in cardiomyocytes. As such, this study establishes a molecular module involving KLF15 and PPARα and provides fundamental insights into the molecular regulation of cardiac lipid metabolism.
Collapse
|
31
|
Fillmore N, Mori J, Lopaschuk GD. Mitochondrial fatty acid oxidation alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br J Pharmacol 2014; 171:2080-90. [PMID: 24147975 DOI: 10.1111/bph.12475] [Citation(s) in RCA: 342] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/20/2013] [Accepted: 09/26/2013] [Indexed: 01/09/2023] Open
Abstract
Heart disease is a leading cause of death worldwide. In many forms of heart disease, including heart failure, ischaemic heart disease and diabetic cardiomyopathies, changes in cardiac mitochondrial energy metabolism contribute to contractile dysfunction and to a decrease in cardiac efficiency. Specific metabolic changes include a relative increase in cardiac fatty acid oxidation rates and an uncoupling of glycolysis from glucose oxidation. In heart failure, overall mitochondrial oxidative metabolism can be impaired while, in ischaemic heart disease, energy production is impaired due to a limitation of oxygen supply. In both of these conditions, residual mitochondrial fatty acid oxidation dominates over mitochondrial glucose oxidation. In diabetes, the ratio of cardiac fatty acid oxidation to glucose oxidation also increases, although primarily due to an increase in fatty acid oxidation and an inhibition of glucose oxidation. Recent evidence suggests that therapeutically regulating cardiac energy metabolism by reducing fatty acid oxidation and/or increasing glucose oxidation can improve cardiac function of the ischaemic heart, the failing heart and in diabetic cardiomyopathies. In this article, we review the cardiac mitochondrial energy metabolic changes that occur in these forms of heart disease, what role alterations in mitochondrial fatty acid oxidation have in contributing to cardiac dysfunction and the potential for targeting fatty acid oxidation to treat these forms of heart disease.
Collapse
Affiliation(s)
- N Fillmore
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | | | | |
Collapse
|
32
|
Santos PP, Oliveira F, Ferreira VCMP, Polegato BF, Roscani MG, Fernandes AA, Modesto P, Rafacho BPM, Zanati SG, Di Lorenzo A, Matsubara LS, Paiva SAR, Zornoff LAM, Minicucci MF, Azevedo PS. The role of lipotoxicity in smoke cardiomyopathy. PLoS One 2014; 9:e113739. [PMID: 25462161 PMCID: PMC4252176 DOI: 10.1371/journal.pone.0113739] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 10/29/2014] [Indexed: 01/09/2023] Open
Abstract
Background/Aims Experimental and clinical studies have shown the direct toxic effects of cigarette smoke (CS) on the myocardium, independent of vascular effects. However, the underlying mechanisms are not well known. Methods Wistar rats were allocated to control (C) and cigarette smoke (CS) groups. CS rats were exposed to cigarette smoke for 2 months. Results After that morphometric, functional and biochemical parameters were measured. The echocardiographic study showed enlargement of the left atria, increase in the left ventricular systolic volume and reduced systolic function. Within the cardiac metabolism, exposure to CS decreased beta hydroxy acyl coenzyme A dehydrogenases and citrate synthases and increased lactate dehydrogenases. Peroxisome proliferator-activated receptor alpha (PPARα) and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) were expressed similarly in both groups. CS increased serum lipids and myocardial triacylglycerols (TGs). These data suggest that impairment in fatty acid oxidation and the accumulation of cardiac lipids characterize lipotoxicity. CS group exhibited increased oxidative stress and decreased antioxidant defense. Finally, the myocyte cross-sectional area and active Caspase 3 were increased in the CS group. Conclusion The cardiac remodeling that was observed in the CS exposure model may be explained by abnormalities in energy metabolism, including lipotoxicity and oxidative stress.
Collapse
Affiliation(s)
- Priscila P. Santos
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Fernando Oliveira
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Vanessa C. M. P. Ferreira
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Bertha F. Polegato
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Meliza G. Roscani
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Ana Angelica Fernandes
- Chemistry and Biochemistry Department, Instituto de Biociências de Botucatu, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Pamela Modesto
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Bruna P. M. Rafacho
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Silmeia G. Zanati
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Center of Vascular Biology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Luiz S. Matsubara
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Sergio A. R. Paiva
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Leonardo A. M. Zornoff
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Marcos F. Minicucci
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Paula S. Azevedo
- Internal Medicine Department, Botucatu Medical School, UNESP - Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
33
|
Wu RM, Sun YY, Zhou TT, Zhu ZY, Zhuang JJ, Tang X, Chen J, Hu LH, Shen X. Arctigenin enhances swimming endurance of sedentary rats partially by regulation of antioxidant pathways. Acta Pharmacol Sin 2014; 35:1274-84. [PMID: 25152028 PMCID: PMC4186987 DOI: 10.1038/aps.2014.70] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/09/2014] [Indexed: 01/01/2023]
Abstract
AIM Arctigenin, a phenylpropanoid dibenzylbutyrolactone lignan found in traditional Chinese herbs, has been determined to exhibit a variety of pharmacological activities, including anti-tumor, anti-inflammation, neuroprotection, and endurance enhancement. In the present study, we investigated the antioxidation and anti-fatigue effects of arctigenin in rats. METHODS Rat L6 skeletal muscle cell line was exposed to H2O2 (700 μmol/L), and ROS level was assayed using DCFH-DA as a probe. Male SD rats were injected with arctigenin (15 mg·kg(-1)·d(-1), ip) for 6 weeks, and then the weight-loaded forced swimming test (WFST) was performed to evaluate their endurance. The levels of antioxidant-related genes in L6 cells and the skeletal muscles of rats were analyzed using real-time RT-PCR and Western blotting. RESULTS Incubation of L6 cells with arctigenin (1, 5, 20 μmol/L) dose-dependently decreased the H2O2-induced ROS production. WFST results demonstrated that chronic administration of arctigenin significantly enhanced the endurance of rats. Furthermore, molecular biology studies on L6 cells and skeletal muscles of the rats showed that arctigenin effectively increased the expression of the antioxidant-related genes, including superoxide dismutase (SOD), glutathione reductase (Gsr), glutathione peroxidase (GPX1), thioredoxin (Txn) and uncoupling protein 2 (UCP2), through regulation of two potential antioxidant pathways: AMPK/PGC-1α/PPARα in mitochondria and AMPK/p53/Nrf2 in the cell nucleus. CONCLUSION Arctigenin efficiently enhances rat swimming endurance by elevation of the antioxidant capacity of the skeletal muscles, which has thereby highlighted the potential of this natural product as an antioxidant in the treatment of fatigue and related diseases.
Collapse
Affiliation(s)
- Ruo-ming Wu
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan-yan Sun
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ting-ting Zhou
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhi-yuan Zhu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing-jing Zhuang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xuan Tang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing Chen
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Li-hong Hu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xu Shen
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
34
|
Lee TI, Kao YH, Chen YC, Tsai WC, Chung CC, Chen YJ. Cardiac metabolism, inflammation, and peroxisome proliferator-activated receptors modulated by 1,25-dihydroxyvitamin D3 in diabetic rats. Int J Cardiol 2014; 176:151-7. [PMID: 25062566 DOI: 10.1016/j.ijcard.2014.07.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/10/2014] [Accepted: 07/05/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND High free fatty acid with reduced glucose utilization in diabetes mellitus (DM) impairs cardiac function. Peroxisome proliferator-activated receptors (PPARs) modulate myocardial lipid and glucose homeostasis. The active 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) regulates oxidative stress and inflammation, which may play a key role in the modulation of PPARs. The aim of this study was to investigate whether 1,25(OH)2D3 can modulate the cardiac PPARs and fatty acid metabolism. METHODS Electrocardiogram, echocardiogram, and Western blot analysis were used to evaluate cardiac fatty acid metabolism, inflammation, and PPAR isoform expression in Wistar-Kyoto (WKY) rats, DM rats, and DM rats treated with 1,25(OH)2D3. RESULTS Compared to healthy rats, DM and 1,25(OH)2D3-treated DM rats had lower body weight. DM rats had larger left ventricular end-diastolic diameter, and longer QT interval than healthy or 1,25(OH)2D3-treated DM rats. Moreover, compared to healthy or 1,25(OH)2D3-treated DM rats, DM rats had fewer cardiac PPAR-α and PPAR-δ protein expressions, but had increased cardiac PPAR-γ protein levels, tumor necrosis factor-α, interleukin-6, 5' adenosine monophosphate-activated protein kinaseα2, phosphorylated acetyl CoA carboxylase, carnitine palmitoyltransferase 1, PPAR-γ coactivator 1-α, cluster of differentiation 36, and diacylglycerol acyltransferase 2 protein expressions. CONCLUSIONS 1,25(OH)2D3 significantly changed the cardiac function and fatty acid regulations in DM hearts, which may be caused by its regulations on cardiac PPARs and proinflammatory cytokines.
Collapse
Affiliation(s)
- Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of General Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chin Tsai
- Division of Cardiology, Tzu-Chi General Hospital, Institute of Medical Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Cheng-Chih Chung
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
35
|
Aung HH, Tsoukalas A, Rutledge JC, Tagkopoulos I. A systems biology analysis of brain microvascular endothelial cell lipotoxicity. BMC SYSTEMS BIOLOGY 2014; 8:80. [PMID: 24993133 PMCID: PMC4112729 DOI: 10.1186/1752-0509-8-80] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/23/2014] [Indexed: 02/08/2023]
Abstract
Background Neurovascular inflammation is associated with a number of neurological diseases including vascular dementia and Alzheimer’s disease, which are increasingly important causes of morbidity and mortality around the world. Lipotoxicity is a metabolic disorder that results from accumulation of lipids, particularly fatty acids, in non-adipose tissue leading to cellular dysfunction, lipid droplet formation, and cell death. Results Our studies indicate for the first time that the neurovascular circulation also can manifest lipotoxicity, which could have major effects on cognitive function. The penetration of integrative systems biology approaches is limited in this area of research, which reduces our capacity to gain an objective insight into the signal transduction and regulation dynamics at a systems level. To address this question, we treated human microvascular endothelial cells with triglyceride-rich lipoprotein (TGRL) lipolysis products and then we used genome-wide transcriptional profiling to obtain transcript abundances over four conditions. We then identified regulatory genes and their targets that have been differentially expressed through analysis of the datasets with various statistical methods. We created a functional gene network by exploiting co-expression observations through a guilt-by-association assumption. Concomitantly, we used various network inference algorithms to identify putative regulatory interactions and we integrated all predictions to construct a consensus gene regulatory network that is TGRL lipolysis product specific. Conclusion System biology analysis has led to the validation of putative lipid-related targets and the discovery of several genes that may be implicated in lipotoxic-related brain microvascular endothelial cell responses. Here, we report that activating transcription factors 3 (ATF3) is a principal regulator of TGRL lipolysis products-induced gene expression in human brain microvascular endothelial cell.
Collapse
Affiliation(s)
| | | | | | - Ilias Tagkopoulos
- UC Davis Genome Center, University of California, Davis, CA 95616, USA.
| |
Collapse
|
36
|
Energy-regulated molecules maintain young status in the trophocytes and fat cells of old queen honeybees. Biogerontology 2014; 15:389-400. [PMID: 24973265 DOI: 10.1007/s10522-014-9509-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 06/16/2014] [Indexed: 12/13/2022]
Abstract
Queen honeybees (Apis mellifera) have much longer lifespans than worker bees. Energy-regulated molecules in the trophocytes and fat cells of workers during aging have been determined, but are unknown in queen bees. In the present study, energy-regulated molecules were evaluated in the trophocytes and fat cells of young and old queen bees. Adenosine monophosphate-activated protein kinase α2 (AMPK-α2), phosphorylated AMPK-α2 (pAMPK-α2), and cAMP-specific phosphodiesterases activity increased with aging. The pAMPK-α2/AMPK-α2 ratio and AMPK activity; adenosine triphosphate (ATP), adenosine diphosphate (ADP), and adenosine monophosphate (AMP) concentrations; the ADP/ATP ratio and the AMP/ATP ratio; the cyclic adenosine monophosphate concentration; forkhead box protein O expression; Silent information regulator T1 (SirT1) expression and activity; and peroxisome proliferator-activated receptor-α (PPAR-α) expression were not significantly different between young and old queen bees. These results show that energy-regulated molecules maintain a youthful status in the trophocytes and fat cells of queen bees during aging. These cells seem to have longevity-promoting mechanisms and may clarify the secret of longevity in queen bees.
Collapse
|
37
|
The PPARβ/δ agonist GW0742 modulates signaling pathways associated with cardiac myocyte growth via a non-genomic redox mechanism. Mol Cell Biochem 2014; 395:145-54. [PMID: 24939361 DOI: 10.1007/s11010-014-2120-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 06/02/2014] [Indexed: 12/28/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the nuclear hormone receptor superfamily and appear to have beneficial effects in the cardiovascular system. PPARβ/δ has been shown previously to exert an inhibitory effect on cardiac myocyte hypertrophy in vivo and in vitro although the exact mechanism is not fully clear yet. The principal signaling pathways that have been involved in triggering cardiac hypertrophic response are mitogen-activated protein kinases (MAPKs) and PI3K/Akt cascades. In this study, we sought to evaluate the potential effects evoked by PPARβ/δ activation on signaling pathways that are implicated in cardiac myocyte growth responses. The selective PPARβ/δ agonist GW0742 attenuated ERK1/2 and Akt phosphorylation that was stimulated by growth promoting agonists (phenylephrine, insulin or IGF-1). This effect was not reversed by the specific PPARβ/δ antagonist, GSK0660, but was inhibited by vanadate, a potent protein tyrosine phosphatase inhibitor. In addition, GW0742 prevented the oxidation and inactivation of PTEN supporting further the notion that its inhibitory action on the agonist-induced kinase phosphorylation is mediated by the modulation of phosphatase activity. Furthermore, GW0742 abolished the agonist-induced intracellular generation of reactive oxygen species, independently of PPARβ/δ activation. Our data reveals a new non-genomic mechanism of GW0742, which ameliorates the generation of reactive oxygen species and attenuates ERK1/2 and PI3K/Akt signaling, with implications in the regulation of cardiac hypertrophic response.
Collapse
|
38
|
Wang L, Wang Y, Liang Y, Li J, Liu Y, Zhang J, Zhang A, Fu J, Jiang G. PFOS induced lipid metabolism disturbances in BALB/c mice through inhibition of low density lipoproteins excretion. Sci Rep 2014; 4:4582. [PMID: 24694979 PMCID: PMC3974142 DOI: 10.1038/srep04582] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 03/19/2014] [Indexed: 12/24/2022] Open
Abstract
Male BALB/c mice fed with either a regular or high fat diet were exposed to 0, 5 or 20 mg/kg perfluorooctane sulfonate (PFOS) for 14 days. Increased body weight, serum glucose, cholesterol and lipoprotein levels were observed in mice given a high fat diet. However, all PFOS-treated mice got reduced levels of serum lipid and lipoprotein. Decreasing liver glycogen content was also observed, accompanied by reduced serum glucose levels. Histological and ultrastructural examination detected more lipid droplets accumulated in hepatocytes after PFOS exposure. Moreover, transcripitonal activity of lipid metabolism related genes suggests that PFOS toxicity is probably unrelevant to PPARα's transcription. The present study demonstrates a lipid disturbance caused by PFOS and thus point to its role in inhibiting the secretion and normal function of low density lipoproteins.
Collapse
Affiliation(s)
- Ling Wang
- 1] State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, P. O. Box 2871, Beijing 100085, P. R. China [2] Key Laboratory of Subtropical Agriculture and Environment, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, P. R. China
| | - Yu Wang
- 1] Key Laboratory of Subtropical Agriculture and Environment, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, P. R. China [2] Department of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, P. R. China
| | - Yong Liang
- 1] School of Medicine, Jianghan University, Wuhan 430056, P. R. China [2] Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, P. R. China
| | - Jia Li
- School of Medicine, Jianghan University, Wuhan 430056, P. R. China
| | - Yuchen Liu
- School of Medicine, Jianghan University, Wuhan 430056, P. R. China
| | - Jie Zhang
- Key Laboratory of Subtropical Agriculture and Environment, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, P. R. China
| | - Aiqian Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, P. O. Box 2871, Beijing 100085, P. R. China
| | - Jianjie Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, P. O. Box 2871, Beijing 100085, P. R. China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, P. O. Box 2871, Beijing 100085, P. R. China
| |
Collapse
|
39
|
Chang PC, Chen LJ, Cheng JT. Role of peroxisome proliferator-activated receptors δ (PPARδ) in rats showing endotoxemic heart failure. J Appl Biomed 2014. [DOI: 10.1016/j.jab.2013.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
40
|
Merit of ginseng in the treatment of heart failure in type 1-like diabetic rats. BIOMED RESEARCH INTERNATIONAL 2014; 2014:484161. [PMID: 24745017 PMCID: PMC3976851 DOI: 10.1155/2014/484161] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/06/2014] [Indexed: 12/30/2022]
Abstract
The present study investigated the merit of ginseng in the improvement of heart failure in diabetic rats and the role of peroxisome proliferator-activated receptors δ (PPAR δ ). We used streptozotocin-induced diabetic rat (STZ-rat) to screen the effects of ginseng on cardiac performance and PPAR δ expression. Changes of body weight, water intake, and food intake were compared in three groups of age-matched rats; the normal control (Wistar rats) received vehicle, STZ-rats received vehicle and ginseng-treated STZ-rats. We also determined cardiac performances in addition to blood glucose level in these animals. The protein levels of PPAR δ in hearts were identified using Western blotting analysis. In STZ-rats, cardiac performances were decreased but the food intake, water intake, and blood glucose were higher than the vehicle-treated control. After a 7-day treatment of ginseng in STZ-rats, cardiac output was markedly enhanced without changes in diabetic parameters. This treatment with ginseng also increased the PPAR δ expression in hearts of STZ-rats. The related signal of cardiac contractility, troponin I phosphorylation, was also raised. Ginseng-induced increasing of cardiac output was reversed by the cotreatment with PPAR δ antagonist GSK0660. Thus, we suggest that ginseng could improve heart failure through the increased PPAR δ expression in STZ-rats.
Collapse
|
41
|
Ginseng is useful to enhance cardiac contractility in animals. BIOMED RESEARCH INTERNATIONAL 2014; 2014:723084. [PMID: 24689053 PMCID: PMC3932289 DOI: 10.1155/2014/723084] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 12/25/2013] [Indexed: 12/24/2022]
Abstract
Ginseng has been shown to be effective on cardiac dysfunction. Recent evidence has highlighted the mediation of peroxisome proliferator-activated receptors (PPARs) in cardiac function. Thus, we are interested to investigate the role of PPARδ in ginseng-induced modification of cardiac contractility. The isolated hearts in Langendorff apparatus and hemodynamic analysis in catheterized rats were applied to measure the actions of ginseng ex vivo and in vivo. In normal rats, ginseng enhanced cardiac contractility and hemodynamic dP/dt(max) significantly. Both actions were diminished by GSK0660 at a dose enough to block PPARδ. However, ginseng failed to modify heart rate at the same dose, although it did produce a mild increase in blood pressure. Data of intracellular calcium level and Western blotting analysis showed that both the PPARδ expression and troponin I phosphorylation were raised by ginseng in neonatal rat cardiomyocyte. Thus, we suggest that ginseng could enhance cardiac contractility through increased PPARδ expression in cardiac cells.
Collapse
|
42
|
Ding Y, Yang KD, Yang Q. The role of PPARδ signaling in the cardiovascular system. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:451-73. [PMID: 24373246 DOI: 10.1016/b978-0-12-800101-1.00014-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARα, β/δ, and γ), members of the nuclear receptor transcription factor superfamily, play important roles in the regulation of metabolism, inflammation, and cell differentiation. All three PPAR subtypes are expressed in the cardiovascular system with various expression patterns. Among the three PPAR subtypes, PPARδ is the least studied but has arisen as a potential therapeutic target for cardiovascular and many other diseases. It is known that PPARδ is ubiquitously expressed and abundantly expressed in cardiomyocytes. Accumulated evidence illustrates the role of PPARδ in regulating cardiovascular function and determining pathological progression. In this chapter, we will discuss the current knowledge in the role of PPARδ in the cardiovascular system, the mechanistic insights, and the potential therapeutic utilization for treating cardiovascular disease.
Collapse
Affiliation(s)
- Yishu Ding
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kevin D Yang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qinglin Yang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
43
|
Hussain A, Nookaew I, Khoomrung S, Andersson L, Larsson I, Hulthén L, Jansson N, Jakubowicz R, Nilsson S, Sandberg AS, Nielsen J, Holmäng A. A maternal diet of fatty fish reduces body fat of offspring compared with a maternal diet of beef and a post-weaning diet of fish improves insulin sensitivity and lipid profile in adult C57BL/6 male mice. Acta Physiol (Oxf) 2013; 209:220-34. [PMID: 23746286 DOI: 10.1111/apha.12130] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/20/2013] [Accepted: 05/31/2013] [Indexed: 01/22/2023]
Abstract
AIM The maternal diet during pregnancy and lactation may affect the long-term health of the offspring. Our aim was to study how a fish or meat diet perinatal and after weaning affects body composition, insulin sensitivity and the profile of n-3 and n-6 polyunsaturated fatty acids (PUFAs) in breast milk, fat depots, skeletal muscle and liver in male adult mice offspring. METHODS During gestation and lactation, C57BL/6 dams were fed a herring- or beef-based diet. Half of the pups in each group changed diets after weaning. In offspring, body composition measured by DEXA, plasma lipid profile and insulin sensitivity measured by euglycemic clamp or QUICKI were monitored to adulthood. Analysis of total FAs by GC-MS were performed in the diet, breast milk and in different tissues. RESULTS At 9 week of age, offspring of herring-fed dams had less body fat than offspring of beef-fed dams. Mice fed herring after weaning had increased insulin sensitivity at 15 week of age, reduced total plasma cholesterol and triglyceride levels, and compared with beef-fed mice, larger interscapular brown adipose tissue depots. The FA composition of the maternal diet was mirrored in breast milk, and the herring diet significantly affected the FA profile of different tissues, leading to an increased content of n-3 PUFAs. CONCLUSION A herring-based maternal diet reduces body fat in the offspring, but the insulin sensitivity, plasma lipids and amount of brown adipose tissue are affected by the offspring's own diet; the herring diet is more beneficial than the beef diet.
Collapse
Affiliation(s)
- A. Hussain
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| | - I. Nookaew
- Department of Chemical and Biological Engineering; Systems Biology; Chalmers University of Technology; Gothenburg; Sweden
| | - S. Khoomrung
- Department of Chemical and Biological Engineering; Systems Biology; Chalmers University of Technology; Gothenburg; Sweden
| | - L. Andersson
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| | - I. Larsson
- Department of Endocrinology, Diabetology and Metabolism; Sahlgrenska University Hospital; Gothenburg; Sweden
| | - L. Hulthén
- Department of Internal Medicine and Clinical Nutrition; Institute of Medicine; The Sahlgrenska Academy, University of Gothenburg; Gothenburg; Sweden
| | - N. Jansson
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| | - R. Jakubowicz
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| | - S. Nilsson
- Department of Mathematical Statistics; Chalmers University of Technology; Gothenburg; Sweden
| | - A.-S. Sandberg
- Department of Chemical and Biological Engineering; Food Science; Chalmers University of Technology; Gothenburg; Sweden
| | - J. Nielsen
- Department of Chemical and Biological Engineering; Systems Biology; Chalmers University of Technology; Gothenburg; Sweden
| | - A. Holmäng
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy; University of Gothenburg; Gothenburg; Sweden
| |
Collapse
|
44
|
Hsu CY, Chuang YL. Changes in energy-regulated molecules in the trophocytes and fat cells of young and old worker honeybees (Apis mellifera). J Gerontol A Biol Sci Med Sci 2013; 69:955-64. [PMID: 24149426 DOI: 10.1093/gerona/glt163] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Trophocytes and fat cells of honeybees (Apis mellifera) have been used for cellular senescence studies, but the changes in the expression, concentration, and activity of cellular energy-regulated molecules that occur with aging in worker bees is unknown. In this study, energy-regulated molecules were evaluated in the trophocytes and fat cells of young and old workers. The results showed that (i) adenosine monophosphate-activated protein kinase-α2 (AMPK-α2) expression increased with aging, whereas phosphorylated AMPK-α2 expression, the phosphorylated AMPK/AMPK ratio, and AMPK activity decreased with aging; (ii) adenosine diphosphate and adenosine triphosphate concentrations decreased with aging, the AMP concentration was unchanged, the adenosine diphosphate/adenosine triphosphate ratio did not change with aging, and the AMP/adenosine triphosphate ratio increased with aging; (iii) the cyclic AMP concentration decreased with aging, and cyclic AMP-specific phosphodiesterases activity increased with aging; (iv) silent information regulator 2 (Sir2) expression increased with aging, whereas its activity decreased with aging; and (v) peroxisome proliferator-activated receptor-α expression decreased with aging. These results show that the trophocytes and fat cells of young workers have higher cellular energy status and express higher levels of energy-regulated molecules than those of old workers and that aging results in a decline in the energy status of trophocytes and fat cells in worker honeybees.
Collapse
Affiliation(s)
- Chin-Yuan Hsu
- Department of Biomedical Sciences and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| | - Yu-Lung Chuang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| |
Collapse
|
45
|
Lee TI, Kao YH, Chen YC, Huang JH, Hsu MI, Chen YJ. The dipeptidyl peptidase-4 inhibitor-sitagliptin modulates calcium dysregulation, inflammation, and PPARs in hypertensive cardiomyocytes. Int J Cardiol 2013; 168:5390-5. [PMID: 24012160 DOI: 10.1016/j.ijcard.2013.08.051] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 08/08/2013] [Accepted: 08/18/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Hypertension induces cardiac dysfunction, calcium (Ca(2+)) dysregulation, and arrhythmogenesis. Dipeptidyl peptidase (DPP)-4 inhibitors, an antidiabetic agent with anti-inflammation and anti-hypertension potential, may regulate peroxisome proliferator-activated receptors (PPARs)-α, -γ, and -δ and Ca(2+) homeostasis. OBJECTIVE The purpose of this study was to investigate whether DPP-4 inhibitor, sitagliptin, can modulate PPARs and Ca(2+) handling proteins in hypertensive hearts. METHODS A Western blot analysis was used to evaluate protein expressions of myocardial PPAR isoforms, tumor necrosis factor (TNF)-α, interleukin (IL)-6, sarcoplasmic reticulum ATPase (SERCA2a), Na(+)-Ca(2+) exchanger (NCX), ryanodine receptor (RyR), voltage-dependent Ca(2+) (CaV1.2), slow-voltage potassium currents (Kvs), angiotensin II type 1 receptor (AT1R), and receptor of advanced glycated end-products (RAGE) from Wistar-Kyoto (WKY) rats, spontaneously hypertensive rats (SHR), and SHR treated with sitagliptin (10mg/kg for 4weeks). Conventional microelectrodes were used to record action potentials (APs) in the ventricular myocytes from each group. RESULTS Compared to the control group, SHR had lower cardiac PPAR-α and PPAR-δ protein expressions, but had greater cardiac PPAR-γ levels, and TNF-α, IL-6, RAGE, and AT1R protein expressions, which were ameliorated in the sitagliptin-treated SHR. SHR had prolonged QT interval and AP duration with less SERCA2a and RyR, and greater CaV1.2 expressions, which were also attenuated in sitagliptin-treated SHR. CONCLUSIONS Sitagliptin significantly changed the cardiac electrophysiological characteristics and Ca(2+) regulation, which may have been caused by its effects on cardiac PPARs, proinflammatory cytokines, and AT1R.
Collapse
Affiliation(s)
- T-I Lee
- Department of General Medicine, College of Medicine, Taipei Medical University, Taiwan; Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Barlaka E, Ledvényiová V, Galatou E, Ferko M, Čarnická S, Ravingerová T, Lazou A. Delayed cardioprotective effects of WY-14643 are associated with inhibition of MMP-2 and modulation of Bcl-2 family proteins through PPAR-α activation in rat hearts subjected to global ischaemia–reperfusion. Can J Physiol Pharmacol 2013; 91:608-16. [DOI: 10.1139/cjpp-2012-0412] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors regulating cardiac lipid metabolism and energy homeostasis. Although the activation of PPARs has been implicated in cardioprotection, the molecular mechanisms are largely unexplored. In this study, we aimed to investigate the effect of the PPAR-α agonist WY-14643 (WY), mimicking a delayed effect of preconditioning in rat hearts exposed to acute ischaemia–reperfusion (I/R) 24 h later, and to define whether antioxidative and antiapoptotic mechanisms are involved. Treatment with WY markedly attenuated post-ischaemic contractile dysfunction (as evidenced by the reduced infarct size), the higher left ventricular developed pressure (LVDP) recovery, and the decreased occurrence of arrhythmias. These effects were abolished in the presence of the PPAR-α antagonist MK886. Heme oxygenase-1, a key antioxidative enzyme implicated in cytoprotection, was upregulated in response to WY at baseline, but was markedly reduced after I/R, indicating reduced oxidative stress. WY treatment was also associated with decreased mRNA levels and enzymatic activity of matrix metalloproteinase-2, and increased ratios of Bcl-2:Bax proteins. These results indicate that PPAR-α activation by its selective ligand WY may confer delayed preconditioning-like protection in rat hearts subjected to I/R by modulating oxidative stress, activation of matrix metalloproteinase-2, and expression of Bcl-2 and Bax.
Collapse
Affiliation(s)
- Eleftheria Barlaka
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Veronika Ledvényiová
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Eleftheria Galatou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Miroslav Ferko
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Slávka Čarnická
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| |
Collapse
|
47
|
Cernecka H, Ochodnicka-Mackovicova K, Kucerova D, Kmecova J, Nemcekova V, Doka G, Kyselovic J, Krenek P, Ochodnicky P, Klimas J. Enalaprilat increases PPARβ/δ expression, without influence on PPARα and PPARγ, and modulate cardiac function in sub-acute model of daunorubicin-induced cardiomyopathy. Eur J Pharmacol 2013; 714:472-7. [PMID: 23834779 DOI: 10.1016/j.ejphar.2013.06.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 06/17/2013] [Accepted: 06/21/2013] [Indexed: 11/28/2022]
Abstract
Anthracycline therapy is limited by a cardiotoxicity that may eventually lead to chronic heart failure which is thought to be prevented by ACE inhibitors (ACEi). However, the protective effect of ACEi in early stages of this specific injury remains elusive. Activated nuclear transcription factors peroxisome proliferator-activated receptors (PPAR) regulate cellular metabolism, but their involvement in anthracycline cardiomyopathy has not been investigated yet. For this purpose, Wistar rats were administered with daunorubicin (i.p., 3 mg/kg, in 48 h intervals) or co-administered with daunorubicine and enalaprilat (i.p., 5 mg/kg in 12 h intervals). Control animals received vehicle. Left ventricular function was measured invasively under anesthesia. Cell-shortening was measured by videomicroscopy in isolated cardiomyocytes. Expression of PPARs mRNA in cardiac tissue was measured by Real-Time PCR. Although the hemodynamic parameters of daunorubicin-treated rats remained altered upon ACEi co-administration, ACEi normalized daunorubicin-induced QT prolongation. On cellular level, ACEi normalized altered basal and isoproterenol-stimulated cardiac cell shortening in daunorubicine-treated group. Moreover, anthracycline administration significantly up-regulated heart PPARα mRNA and its expression remained increased after ACEi co-administration. On the other hand, the expression of cardiac PPARβ/δ was not altered in anthracycline-treated animals, whereas co-administration of ACEi increased its expression. Conclusively, effect of ACEi can be already detected in sub-acute phase of anthracycline-induced cardiotoxicity. Altered expression of heart PPARs may suggest these nuclear receptors as a novel target in anthracycline cardiomyopathy.
Collapse
Affiliation(s)
- Hana Cernecka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lee TI, Kao YH, Chen YC, Huang JH, Hsiao FC, Chen YJ. Peroxisome proliferator-activated receptors modulate cardiac dysfunction in diabetic cardiomyopathy. Diabetes Res Clin Pract 2013; 100:330-9. [PMID: 23369225 DOI: 10.1016/j.diabres.2013.01.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/23/2012] [Accepted: 01/03/2013] [Indexed: 02/08/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality among patients with diabetes mellitus (DM). Chronic inflammation and derangement of myocardial energy and lipid homeostasis are common features of DM. The transcription factors of peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor superfamily, which are important in regulating energy and lipid homeostasis. There are three PPAR isoforms, α, γ, and δ, and their roles have been increasingly recognized to be important in CVD. These three isoforms are expressed in the heart and play pivotal roles in myocardial lipid metabolism, as well as glucose and energy homeostasis, and contribute to extra metabolic roles with effects on inflammation and oxidative stress. Moreover, regulation of PPARs may have significant effects on cardiac electrical activity and arrhythmogenesis. This review describes the roles of PPARs and their agonists in DM cardiomyopathy, inflammation, and cardiac electrophysiology.
Collapse
Affiliation(s)
- T-I Lee
- Department of General Medicine, College of Medicine, Taipei Medical University, Taiwan
| | | | | | | | | | | |
Collapse
|
49
|
Chen ZC, Lee KS, Chen LJ, Wang LY, Niu HS, Cheng JT. Cardiac peroxisome proliferator-activated receptor δ (PPARδ) as a new target for increased contractility without altering heart rate. PLoS One 2013; 8:e64229. [PMID: 23724037 PMCID: PMC3665891 DOI: 10.1371/journal.pone.0064229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 04/12/2013] [Indexed: 12/31/2022] Open
Abstract
Background and Aims Agents having a positive inotropic effect on the heart are widely used for the treatment of heart failure. However, these agents have the side effect of altering heart rate. It has been established that peroxisome proliferator-activated receptor δ (PPARδ) is mediated in cardiac contraction, however the effect on heart rate is unknown. Thus, we used an agonist of PPARδ, GW0742, to investigate this issue in the present study. Methods and Results We used isolated hearts in Langendorff apparatus and hemodynamic analysis in catheterized rats to measure the actions of GW0742 extra-vivo and in vivo. In diabetic rats with heart failure, GW0742 at a dose sufficient to activate PPARδ reversed cardiac contraction without changes in heart rate. In normal rats, PPARδ enhanced cardiac contractility and hemodynamic dP/dtmax significantly more than dobutamine. Both actions were diminished by GSK0660 at a dose enough to block PPARδ. However, GW0742 at the same dose failed to modify heart rate, although it did produce a mild increase in blood pressure. Detection of intracellular calcium level and Western blotting analysis showed that the intracellular calcium concentration and troponin I phosphorylation were both enhanced by GW0742. Conclusion Activation of PPARδ by GW0742 increases cardiac contractility but not heart rate. Thus, PPARδ may be a suitable target for the development of inotropic agents to treat heart failure without changing heart rate.
Collapse
Affiliation(s)
- Zhih-Cherng Chen
- Department of Cardiology, Chi-Mei Medical Center, Yong Kang, Tainan City, Taiwan
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy & Science, Jean-Tae, Tainan City, Taiwan
| | - Kung Shing Lee
- Department of Surgery, Kaohsiung Municipal Hsiao-Kang Hospital, and Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Li-Jen Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Lin-Yu Wang
- Department of Pediatrics, Chi-Mei Medical Center, Yong Kang, Tainan City, Taiwan
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi College of Technology, Hualien City, Taiwan
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- * E-mail:
| |
Collapse
|
50
|
Lo SH, Lee KS, Chen LJ, Cheng JT, Chen CH. Increase of PPARδ by dopamine mediated via DA-1 receptor-linked phospholipase C pathway in neonatal rat cardiomyocytes. Auton Neurosci 2013; 177:211-6. [PMID: 23701913 DOI: 10.1016/j.autneu.2013.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 04/29/2013] [Accepted: 04/30/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND Role of peroxisome proliferator-activated receptor δ (PPARδ) in cardiac contraction has recently been established. Dopamine is one of the agents used to treat heart failure in clinics. But the mediation of PPARδ in cardiac action of dopamine is still unclear. METHODS The present study is aimed to clarify this point using neonatal rat cardiomyocytes to investigate the changes of PPARδ expression and cardiac troponin I (cTnI) phosphorylation by Western blotting analysis. Antagonists of receptors, inhibitor of phospholipase C (PLC) (U73122), calcium chelator (BAPTA-AM), and inhibitor of protein kinase A (PKAI) were also applied. We silenced PPARδ by RNAi to identify the major role of PPARδ in dopamine-induced actions. RESULTS Dopamine increases PPARδ expression and cardiac troponin I (cTnI) phosphorylation in a time- and dose-dependent manner in neonatal rat cardiomyocytes. Moreover, both actions of dopamine were blocked by DA1 receptor antagonist and PLC inhibitor but not by PKAI. The increase of cTnI phosphorylation by dopamine was also inhibited in cardiomyocytes silenced by RNAi of PPARδ. CONCLUSION We suggest that dopamine can enhance cardiac contraction mainly through an activation of DA1 receptor-linked PLC pathway to increase cellular calcium ions for the increase of PPARδ expression.
Collapse
Affiliation(s)
- Shih-Hsiang Lo
- Department of Internal Medicine - Cardiology, Taipei City Hospital - Zhongxing Branch, Datong Dist., Taipei City 10341, Taiwan
| | | | | | | | | |
Collapse
|