1
|
Jiang W, Liu L, Wang M, Li X, Zhou T, Hou X, Qiao L, Chen C, Zuo D, Liu J, Ren L. KLF15 suppresses stemness of pancreatic cancer by decreasing USP21-mediated Nanog stability. Cell Mol Life Sci 2024; 81:417. [PMID: 39367978 PMCID: PMC11455850 DOI: 10.1007/s00018-024-05442-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/08/2024] [Accepted: 08/22/2024] [Indexed: 10/07/2024]
Abstract
The existence of cancer stem cells (CSCs) in pancreatic ductal adenocarcinoma (PDAC) is considered to be the key factor for metastasis and chemoresistance. Thus, novel therapeutic strategies for eradicating CSCs are urgently needed. Here we aimed to explore the role of KLF15 in stemness and the feasibility of using KLF15 to inhibit CSCs and improve chemotherapy sensitivity in PDAC. In this study, we report that KLF15 is negatively associated with poor survival and advanced pathological staging of PDAC. Moreover, tumorous KLF15 suppresses the stemness of PDAC by promoting the degradation of Nanog, and KLF15 directly interacts with Nanog, inhibiting interaction between Nanog with USP21. We also demonstrate that the KLF15/Nanog complex inhibit the stemness in vivo and in PDX cells. Tazemetostat suppresses stemness and sensitizes PDAC cells to gemcitabine by promoting KLF15 expression in PDAC. In summary, the findings of our study confirm the value of KLF15 level in diagnosis and prognosis of PDAC, it is the first time to explore the inhibition role of KLF15 in stemness of PDAC and the regulation mechanism of Nanog, contributing to provide a new therapeutic strategy that using Tazemetostat sensitizes PDAC cells to gemcitabine by promoting KLF15 expression for PDAC.
Collapse
Affiliation(s)
- Wenna Jiang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Lin Liu
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Meng Wang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Xueyang Li
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Tianxing Zhou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xupeng Hou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Lu Qiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences of Tianjin Medical University, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Duo Zuo
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Jing Liu
- Department of Breast Oncoplastic Surgery, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Li Ren
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China.
| |
Collapse
|
2
|
Chou MY, Lee CH, Hsieh PL, Chao SC, Yu CH, Liao YW, Lee SP, Yu CC, Fan JY. Targeting microRNA-190a halts the persistent myofibroblast activation and oxidative stress accumulation through upregulation of Krüppel-like factor 15 in oral submucous fibrosis. J Dent Sci 2024; 19:1999-2006. [PMID: 39347084 PMCID: PMC11437311 DOI: 10.1016/j.jds.2024.07.002] [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: 06/14/2024] [Revised: 07/02/2024] [Indexed: 10/01/2024] Open
Abstract
Background/purpose Oral submucous fibrosis (OSF) is a condition characterized by inflammation and excessive collagen deposition, which has been identified as a potentially malignant disorder. Recently, several microRNAs (miRNAs) have been shown to be implicated in various disorders associated with fibrosis. However, how these miRNAs modulate OSF development is poorly understood. Therefore, the study aimed to identify the specific miRNAs that contribute to the progression of OSF and to investigate their molecular mechanisms in promoting fibrosis. Materials and methods The expression and clinical significance of potential pro-fibrosis miRNA in the OSF cohort and primary buccal mucosal fibroblasts were confirmed through RNA sequencing and qRT-PCR. Luciferase reporter activity assay, miRNA mimic or inhibitor, and short-hairpin RNA silencing were used to elucidate the molecular mechanism of miRNA. Transwell migration, collagen contraction, and reactive oxygen species (ROS) generation detection were used to investigate the effects of this mechanism on the myofibroblast phenotype and cellular pro-fibrosis capacity. Results This study demonstrated that miR-190a was overexpressed in fibrotic buccal mucosal fibroblasts (fBMFs). Transfecting fBMFs with miR-190a inhibitor resulted in reduced cell migration, collagen gel contraction, ROS generation, and expression of fibrotic markers. Furthermore, miR-190a exerted this pro-fibrosis property by direct binding to its target, Krüppel-like factor 15 (KLF15). The results also indicated that the aberrant upregulation of miR-190a, in turn, downregulated the expression of KLF15, which resulted in the activation of myofibroblast. Conclusion Our findings demonstrated that miR-190a was involved in myofibroblast activation, suggesting that targeting the miR-190a/KLF15 axis may be a feasible approach in the therapy of OSF.
Collapse
Affiliation(s)
- Ming-Yung Chou
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Hsuan Lee
- Department of Dentistry, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Chi Chao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chuan-Hang Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Wen Liao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shiao-Pieng Lee
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Tri-Service General Hospital, Taipei, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Jun-Yang Fan
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
3
|
Durumutla HB, Prabakaran AD, Soussi FEA, Akinborewa O, Latimer H, McFarland K, Piczer K, Werbrich C, Jain MK, Haldar SM, Quattrocelli M. Glucocorticoid chrono-pharmacology unveils novel targets for the cardiomyocyte-specific GR-KLF15 axis in cardiac glucose metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572210. [PMID: 38187555 PMCID: PMC10769285 DOI: 10.1101/2023.12.18.572210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Circadian time-of-intake gates the cardioprotective effects of glucocorticoid administration in both healthy and infarcted hearts. The cardiomyocyte-specific glucocorticoid receptor (GR) and its co-factor, Krüppel-like factor (Klf15), play critical roles in maintaining normal heart function in the long-term and serve as pleiotropic regulators of cardiac metabolism. Despite this understanding, the cardiomyocyte-autonomous metabolic targets influenced by the concerted epigenetic action of GR-Klf15 axis remain undefined. Here, we demonstrate the critical roles of the cardiomyocyte-specific GR and Klf15 in orchestrating a circadian-dependent glucose oxidation program within the heart. Combining integrated transcriptomics and epigenomics with cardiomyocyte-specific inducible ablation of GR or Klf15, we identified their synergistic role in the activation of adiponectin receptor expression (Adipor1) and the mitochondrial pyruvate complex (Mpc1/2), thereby enhancing insulin-stimulated glucose uptake and pyruvate oxidation. Furthermore, in obese diabetic (db/db) mice exhibiting insulin resistance and impaired glucose oxidation, light-phase prednisone administration, as opposed to dark-phase prednisone dosing, effectively restored cardiomyocyte glucose oxidation and improved diastolic function towards control-like levels in a sex-independent manner. Collectively, our findings uncover novel cardiomyocyte-autonomous metabolic targets of the GR-Klf15 axis. This study highlights the circadian-dependent cardioprotective effects of glucocorticoids on cardiomyocyte glucose metabolism, providing critical insights into chrono-pharmacological strategies for glucocorticoid therapy in cardiovascular disease.
Collapse
Affiliation(s)
- Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ashok Daniel Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Olukunle Akinborewa
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mukesh K Jain
- Dept Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Saptarsi M Haldar
- Amgen Research, South San Francisco, CA, USA; Gladstone Institutes, San Francisco, CA, USA and Dept Medicine, Cardiology Division, UCSF, San Francisco, CA, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
4
|
Choi BH, Hyun S, Koo SH. The role of BCAA metabolism in metabolic health and disease. Exp Mol Med 2024; 56:1552-1559. [PMID: 38956299 PMCID: PMC11297153 DOI: 10.1038/s12276-024-01263-6] [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: 10/28/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 07/04/2024] Open
Abstract
It has long been postulated that dietary restriction is beneficial for ensuring longevity and extending the health span of mammals, including humans. In particular, a reduction in protein consumption has been shown to be specifically linked to the beneficial effect of dietary restriction on metabolic disorders, presumably by reducing the activity of the mechanistic target of rapamycin complex (mTORC) 1 and the reciprocal activation of AMP-activated protein kinase (AMPK) and sirtuin pathways. Although it is widely used as a dietary supplement to delay the aging process in humans, recent evidence suggests that branched-chain amino acids (BCAAs) might be a major cause of the deteriorating effect of a protein diet on aging and related disorders. In this review, we delineate the regulation of metabolic pathways for BCAAs at the tissue-specific level and summarize recent findings regarding the role of BCAAs in the control of metabolic health and disease in mammals.
Collapse
Affiliation(s)
| | - Seunghoon Hyun
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University, Seoul, Korea.
| |
Collapse
|
5
|
Wu HY, Ji ZH, Xie WY, Guo HX, Zheng Y, Gao W, Yuan B. KLF4 promotes milk fat synthesis by regulating the PI3K-AKT-mTOR pathway and targeting FASN activation in bovine mammary epithelial cells. iScience 2024; 27:109850. [PMID: 38779481 PMCID: PMC11108978 DOI: 10.1016/j.isci.2024.109850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/18/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Milk fat is an important indicator for evaluating the quality of cow's milk. In this study, we used bovine mammary epithelial cells (BMECs) to investigate the role and molecular mechanism of KLF4 in the regulation of milk fat synthesis. The results showed that KLF4 was more highly expressed in mammary tissues of high-fat cows compared with low-fat cows. KLF4 positively regulated the expression of genes related to milk fat synthesis in BMECs, increasing intracellular triglycerides content, and KLF4 promoted milk fat synthesis by activating the PI3K-AKT-mTOR signaling pathway. Furthermore, the results of animal experiments also confirmed that knockdown of KLF4 inhibited milk fat synthesis. In addition, yeast one-hybrid assays and dual-luciferase reporter gene assays confirmed that KLF4 directly targets and binds to the fatty acid synthase (FASN) promoter region to promote FASN transcription. These results demonstrate that KLF4 is a key transcription factor for milk fat synthesis in BMECs.
Collapse
Affiliation(s)
- Hong-Yu Wu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
- Jilin Academy of Agricultural Sciences, Jilin 132101, China
| | - Zhong-Hao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
- Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - Wen-Yin Xie
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Hai-Xiang Guo
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Yi Zheng
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Wei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| |
Collapse
|
6
|
Scicchitano P, Amati F, Ciccone MM, D’Ascenzi F, Imbalzano E, Liga R, Paolillo S, Pastore MC, Rinaldi A, Mattioli AV, Cameli M. Hypertriglyceridemia: Molecular and Genetic Landscapes. Int J Mol Sci 2024; 25:6364. [PMID: 38928071 PMCID: PMC11203941 DOI: 10.3390/ijms25126364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Lipid disorders represent one of the most worrisome cardiovascular risk factors. The focus on the impact of lipids on cardiac and vascular health usually concerns low-density lipoprotein cholesterol, while the role of triglycerides (TGs) is given poor attention. The literature provides data on the impact of higher plasma concentrations in TGs on the cardiovascular system and, therefore, on the outcomes and comorbidities of patients. The risk for coronary heart diseases varies from 57 to 76% in patients with hypertriglyceridemia. Specifically, the higher the plasma concentrations in TGs, the higher the incidence and prevalence of death, myocardial infarction, and stroke. Nevertheless, the metabolism of TGs and the exact physiopathologic mechanisms which try to explain the relationship between TGs and cardiovascular outcomes are not completely understood. The aims of this narrative review were as follows: to provide a comprehensive evaluation of the metabolism of triglycerides and a possible suggestion for understanding the targets for counteracting hypertriglyceridemia; to describe the inner physiopathological background for the relationship between vascular and cardiac damages derived from higher plasma concentrations in TGs; and to outline the need for promoting further insights in therapies for reducing TGs plasma levels.
Collapse
Affiliation(s)
- Pietro Scicchitano
- Cardiology Department, Hospital “F Perinei” ASL BA, 70022 Altamura, Italy
| | - Francesca Amati
- Cardiovascular Diseases Section, University of Bari, 70124 Bari, Italy; (F.A.); (M.M.C.)
| | - Marco Matteo Ciccone
- Cardiovascular Diseases Section, University of Bari, 70124 Bari, Italy; (F.A.); (M.M.C.)
| | - Flavio D’Ascenzi
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy; (F.D.); (M.C.P.); (M.C.)
| | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Riccardo Liga
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy;
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80138 Naples, Italy;
| | - Maria Concetta Pastore
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy; (F.D.); (M.C.P.); (M.C.)
| | - Andrea Rinaldi
- Unit of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Sant’Orsola-Malpighi Hospital, IRCCS, 40138 Bologna, Italy;
| | - Anna Vittoria Mattioli
- Department of Science of Quality of Life, University of Bologna “Alma Mater Studiorum”, 40126 Bologna, Italy;
| | - Matteo Cameli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy; (F.D.); (M.C.P.); (M.C.)
| |
Collapse
|
7
|
Gui LK, Liu HJ, Jin LJ, Peng XC. Krüpple-like factors in cardiomyopathy: emerging player and therapeutic opportunities. Front Cardiovasc Med 2024; 11:1342173. [PMID: 38516000 PMCID: PMC10955087 DOI: 10.3389/fcvm.2024.1342173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024] Open
Abstract
Cardiomyopathy, a heterogeneous pathological condition characterized by changes in cardiac structure or function, represents a significant risk factor for the prevalence and mortality of cardiovascular disease (CVD). Research conducted over the years has led to the modification of definition and classification of cardiomyopathy. Herein, we reviewed seven of the most common types of cardiomyopathies, including Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC), diabetic cardiomyopathy, Dilated Cardiomyopathy (DCM), desmin-associated cardiomyopathy, Hypertrophic Cardiomyopathy (HCM), Ischemic Cardiomyopathy (ICM), and obesity cardiomyopathy, focusing on their definitions, epidemiology, and influencing factors. Cardiomyopathies manifest in various ways ranging from microscopic alterations in cardiomyocytes, to tissue hypoperfusion, cardiac failure, and arrhythmias caused by electrical conduction abnormalities. As pleiotropic Transcription Factors (TFs), the Krüppel-Like Factors (KLFs), a family of zinc finger proteins, are involved in regulating the setting and development of cardiomyopathies, and play critical roles in associated biological processes, including Oxidative Stress (OS), inflammatory reactions, myocardial hypertrophy and fibrosis, and cellular autophagy and apoptosis, particularly in diabetic cardiomyopathy. However, research into KLFs in cardiomyopathy is still in its early stages, and the pathophysiologic mechanisms of some KLF members in various types of cardiomyopathies remain unclear. This article reviews the roles and recent research advances in KLFs, specifically those targeting and regulating several cardiomyopathy-associated processes.
Collapse
Affiliation(s)
- Le-Kun Gui
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Huang-Jun Liu
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Li-Jun Jin
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Xiao-Chun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
- Laboratory of Oncology, School of Basic Medicine, Center for Molecular Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
8
|
Boye C, Kalita CA, Findley AS, Alazizi A, Wei J, Wen X, Pique-Regi R, Luca F. Characterization of caffeine response regulatory variants in vascular endothelial cells. eLife 2024; 13:e85235. [PMID: 38334359 PMCID: PMC10901511 DOI: 10.7554/elife.85235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/08/2024] [Indexed: 02/10/2024] Open
Abstract
Genetic variants in gene regulatory sequences can modify gene expression and mediate the molecular response to environmental stimuli. In addition, genotype-environment interactions (GxE) contribute to complex traits such as cardiovascular disease. Caffeine is the most widely consumed stimulant and is known to produce a vascular response. To investigate GxE for caffeine, we treated vascular endothelial cells with caffeine and used a massively parallel reporter assay to measure allelic effects on gene regulation for over 43,000 genetic variants. We identified 665 variants with allelic effects on gene regulation and 6 variants that regulate the gene expression response to caffeine (GxE, false discovery rate [FDR] < 5%). When overlapping our GxE results with expression quantitative trait loci colocalized with coronary artery disease and hypertension, we dissected their regulatory mechanisms and showed a modulatory role for caffeine. Our results demonstrate that massively parallel reporter assay is a powerful approach to identify and molecularly characterize GxE in the specific context of caffeine consumption.
Collapse
Affiliation(s)
- Carly Boye
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Cynthia A Kalita
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Anthony S Findley
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Adnan Alazizi
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Julong Wei
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Xiaoquan Wen
- Department of Biostatistics, University of MichiganAnn ArborUnited States
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State UniversityDetroitUnited States
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State UniversityDetroitUnited States
- Department of Biology, University of Rome Tor VergataRomeItaly
| |
Collapse
|
9
|
Sakamoto T, Kelly DP. Cardiac maturation. J Mol Cell Cardiol 2024; 187:38-50. [PMID: 38160640 PMCID: PMC10923079 DOI: 10.1016/j.yjmcc.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The heart undergoes a dynamic maturation process following birth, in response to a wide range of stimuli, including both physiological and pathological cues. This process entails substantial re-programming of mitochondrial energy metabolism coincident with the emergence of specialized structural and contractile machinery to meet the demands of the adult heart. Many components of this program revert to a more "fetal" format during development of pathological cardiac hypertrophy and heart failure. In this review, emphasis is placed on recent progress in our understanding of the transcriptional control of cardiac maturation, encompassing the results of studies spanning from in vivo models to cardiomyocytes derived from human stem cells. The potential applications of this current state of knowledge to new translational avenues aimed at the treatment of heart failure is also addressed.
Collapse
Affiliation(s)
- Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Zhai R, Wang W, Zhang D, Li X, Zhang Y, Zhao Y, Zhao L, Wang J, Xu D, Cheng J, Li W, Zhou B, Lin C, Zeng X, Yang X, Ma Z, Liu J, Cui P, Zhang XX. Novel polymorphism at KLF15 gene and its association with growth traits in Hu sheep. Anim Biotechnol 2023; 34:3287-3293. [PMID: 36346056 DOI: 10.1080/10495398.2022.2138413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Growth traits are important economic characteristics of livestock and poultry. In the present study, the expression features of KLF15 and the relationship between KLF15 gene polymorphisms and growth traits in Hu sheep were investigated by using real-time quantitative PCR technology (qPCR), Sanger sequencing, and Kaspar genotyping technology. The qPCR results showed that the KLF15 gene is expressed widely in the tested tissues of Hu sheep, and the expression level of the KLF15 gene in the heart and the muscle was significantly higher than in other tissues (p < 0.05). Missense mutation c.62565119 A > G was found in KLF15, and an association analysis showed that it was correlated with the growth traits (body weight, body height, and body length) of Hu sheep (p < 0.05). The body weight, body height, and body length of the sheep carrying the AA genotype were remarkably higher than those of the GG and AG genotypes (p < 0.05). These results showed that novel polymorphisms at the KLF15 gene can be used as a genetic marker of growth traits of Hu sheep.
Collapse
Affiliation(s)
- Rui Zhai
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Weimin Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Deyin Zhang
- The State Key Laboratory of Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaolong Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yukun Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yuan Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Liming Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Jianghui Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Dan Xu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Jiangbo Cheng
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Wenxin Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Bubo Zhou
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Changchun Lin
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiwen Zeng
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiaobin Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Zongwu Ma
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Jia Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Panpan Cui
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiao Xue Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Liu X, Xu X, Zhang T, Xu L, Tao H, Liu Y, Zhang Y, Meng X. Fatty acid metabolism disorders and potential therapeutic traditional Chinese medicines in cardiovascular diseases. Phytother Res 2023; 37:4976-4998. [PMID: 37533230 DOI: 10.1002/ptr.7965] [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: 04/16/2023] [Revised: 06/13/2023] [Accepted: 07/07/2023] [Indexed: 08/04/2023]
Abstract
Cardiovascular diseases are currently the primary cause of mortality in the whole world. Growing evidence indicated that the disturbances in cardiac fatty acid metabolism are crucial contributors in the development of cardiovascular diseases. The abnormal cardiac fatty acid metabolism usually leads to energy deficit, oxidative stress, excessive apoptosis, and inflammation. Targeting fatty acid metabolism has been regarded as a novel approach to the treatment of cardiovascular diseases. However, there are currently no specific drugs that regulate fatty acid metabolism to treat cardiovascular diseases. Many traditional Chinese medicines have been widely used to treat cardiovascular diseases in clinics. And modern studies have shown that they exert a cardioprotective effect by regulating the expression of key proteins involved in fatty acid metabolism, such as peroxisome proliferator-activated receptor α and carnitine palmitoyl transferase 1. Hence, we systematically reviewed the relationship between fatty acid metabolism disorders and four types of cardiovascular diseases including heart failure, coronary artery disease, cardiac hypertrophy, and diabetic cardiomyopathy. In addition, 18 extracts and eight monomer components from traditional Chinese medicines showed cardioprotective effects by restoring cardiac fatty acid metabolism. This work aims to provide a reference for the finding of novel cardioprotective agents targeting fatty acid metabolism.
Collapse
Affiliation(s)
- Xianfeng Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Xinmei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Tao Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Lei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Honglin Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Yue Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, Sichuan, People's Republic of China
| |
Collapse
|
12
|
Chen X, Shi W, Xie Y, Wang Y, Yao Q, Ke H, Xu X, Liu H, Liu P, Zhou X. Hepatic Krüppel-like factor 14 regulates lipid metabolism in nonalcoholic steatohepatitis mice. FASEB J 2023; 37:e23070. [PMID: 37389939 DOI: 10.1096/fj.202300448r] [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: 03/09/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
Excessive lipid accumulation is a critical characteristic in the development of nonalcoholic steatohepatitis (NASH). The underlying molecular mechanism, however, is unclear. In this study, we explored whether and how Krüppel-like factor 14 (KLF14) affects hepatic lipid metabolism in NASH. KLF14 expression was detected in NASH patients and mice fed a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD). Adeno-associated viruses and adenoviruses were used to alter hepatic KLF14 expression in vivo or in vitro to investigate how KLF14 functions in lipid regulation. The molecular mechanisms were explored using RNA-seq, luciferase reporter, and ChIP assays. The fatty liver phenotype was analyzed histopathologically, and serum and hepatocyte biochemical parameters were measured. The NASH mouse model developed quickly in C57BL/6J mice fed a CDAHFD for 8 weeks. We found that KLF14 expression was decreased in NASH patients and CDAHFD mice. Oleic acid and palmitic acid treatment also reduced KLF14 levels in hepatocytes. KLF14 knockdown downregulated the genes involved in fatty acid oxidation, promoting the progression of hepatic steatosis. In contrast, hepatic KLF14 overexpression alleviated lipid accumulation and oxidative stress in CDAHFD mice. These effects resulted from direct activation of the PPARα signaling pathway. PPARα inhibition diminished the KLF14 overexpression-reduced protective effects against steatosis in OA&PA-treated MPHs and AAV-KLF14-infected CDAHFD mice. These data reveal that hepatic KLF14 regulates lipid accumulation and oxidative stress through the KLF14-PPARα pathway as NASH progresses. KLF14 may be a novel therapeutic target for hepatic steatosis.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Wenjie Shi
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Yunwu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qian Yao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Huajing Ke
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Hui Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, China
| | - Pi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The People's Hospital of Longhua, Shenzhen, China
| | - Xiaojiang Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
13
|
Kumar A, He S, Mali P. Systematic discovery of transcription factors that improve hPSC-derived cardiomyocyte maturation via temporal analysis of bioengineered cardiac tissues. APL Bioeng 2023; 7:026109. [PMID: 37252678 PMCID: PMC10219684 DOI: 10.1063/5.0137458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/09/2023] [Indexed: 05/31/2023] Open
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have the potential to become powerful tools for disease modeling, drug testing, and transplantation; however, their immaturity limits their applications. Transcription factor (TF) overexpression can improve hPSC-CM maturity, but identifying these TFs has been elusive. Toward this, we establish here an experimental framework for systematic identification of maturation enhancing factors. Specifically, we performed temporal transcriptome RNAseq analyses of progressively matured hPSC-derived cardiomyocytes across 2D and 3D differentiation systems and further compared these bioengineered tissues to native fetal and adult-derived tissues. These analyses revealed 22 TFs whose expression did not increase in 2D differentiation systems but progressively increased in 3D culture systems and adult mature cell types. Individually overexpressing each of these TFs in immature hPSC-CMs identified five TFs (KLF15, ZBTB20, ESRRA, HOPX, and CAMTA2) as regulators of calcium handling, metabolic function, and hypertrophy. Notably, the combinatorial overexpression of KLF15, ESRRA, and HOPX improved all three maturation parameters simultaneously. Taken together, we introduce a new TF cocktail that can be used in solo or in conjunction with other strategies to improve hPSC-CM maturation and anticipate that our generalizable methodology can also be implemented to identify maturation-associated TFs for other stem cell progenies.
Collapse
Affiliation(s)
- Aditya Kumar
- Department of Bioengineering, University of California, San Diego, California 92093, USA
| | - Starry He
- Department of Bioengineering, University of California, San Diego, California 92093, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, California 92093, USA
| |
Collapse
|
14
|
Kankuri E, Finckenberg P, Leinonen J, Tarkia M, Björk S, Purhonen J, Kallijärvi J, Kankainen M, Soliymani R, Lalowski M, Mervaala E. Altered acylcarnitine metabolism and inflexible mitochondrial fuel utilization characterize the loss of neonatal myocardial regeneration capacity. Exp Mol Med 2023; 55:806-817. [PMID: 37009793 PMCID: PMC10167339 DOI: 10.1038/s12276-023-00967-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 11/29/2022] [Accepted: 12/23/2022] [Indexed: 04/04/2023] Open
Abstract
Myocardial regeneration capacity declines during the first week after birth, and this decline is linked to adaptation to oxidative metabolism. Utilizing this regenerative window, we characterized the metabolic changes in myocardial injury in 1-day-old regeneration-competent and 7-day-old regeneration-compromised mice. The mice were either sham-operated or received left anterior descending coronary artery ligation to induce myocardial infarction (MI) and acute ischemic heart failure. Myocardial samples were collected 21 days after operations for metabolomic, transcriptomic and proteomic analyses. Phenotypic characterizations were carried out using echocardiography, histology and mitochondrial structural and functional assessments. In both groups, MI induced an early decline in cardiac function that persisted in the regeneration-compromised mice over time. By integrating the findings from metabolomic, transcriptomic and proteomic examinations, we linked regeneration failure to the accumulation of long-chain acylcarnitines and insufficient metabolic capacity for fatty acid beta-oxidation. Decreased expression of the redox-sensitive mitochondrial Slc25a20 carnitine-acylcarnitine translocase together with a decreased reduced:oxidized glutathione ratio in the myocardium in the regeneration-compromised mice pointed to a defect in the redox-sensitive acylcarnitine transport to the mitochondrial matrix. Rather than a forced shift from the preferred adult myocardial oxidative fuel source, our results suggest the facilitation of mitochondrial fatty acid transport and improvement of the beta-oxidation pathway as a means to overcome the metabolic barrier for repair and regeneration in adult mammals after MI and heart failure.
Collapse
Affiliation(s)
- E Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - P Finckenberg
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Leinonen
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Tarkia
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - S Björk
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Purhonen
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Kallijärvi
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Kankainen
- Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Soliymani
- Helsinki Institute of Life Science (HiLIFE), Meilahti Clinical Proteomics Core Facility, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Lalowski
- Helsinki Institute of Life Science (HiLIFE), Meilahti Clinical Proteomics Core Facility, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - E Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
15
|
The KLF7/PFKL/ACADL axis modulates cardiac metabolic remodelling during cardiac hypertrophy in male mice. Nat Commun 2023; 14:959. [PMID: 36810848 PMCID: PMC9944323 DOI: 10.1038/s41467-023-36712-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
The main hallmark of myocardial substrate metabolism in cardiac hypertrophy or heart failure is a shift from fatty acid oxidation to greater reliance on glycolysis. However, the close correlation between glycolysis and fatty acid oxidation and underlying mechanism by which causes cardiac pathological remodelling remain unclear. We confirm that KLF7 simultaneously targets the rate-limiting enzyme of glycolysis, phosphofructokinase-1, liver, and long-chain acyl-CoA dehydrogenase, a key enzyme for fatty acid oxidation. Cardiac-specific knockout and overexpression KLF7 induce adult concentric hypertrophy and infant eccentric hypertrophy by regulating glycolysis and fatty acid oxidation fluxes in male mice, respectively. Furthermore, cardiac-specific knockdown phosphofructokinase-1, liver or overexpression long-chain acyl-CoA dehydrogenase partially rescues the cardiac hypertrophy in adult male KLF7 deficient mice. Here we show that the KLF7/PFKL/ACADL axis is a critical regulatory mechanism and may provide insight into viable therapeutic concepts aimed at the modulation of cardiac metabolic balance in hypertrophied and failing heart.
Collapse
|
16
|
Abstract
Research conducted in the past 15 years has yielded crucial insights that are reshaping our understanding of the systems physiology of branched-chain amino acid (BCAA) metabolism and the molecular mechanisms underlying the close relationship between BCAA homeostasis and cardiovascular health. The rapidly evolving literature paints a complex picture, in which numerous tissue-specific and disease-specific modes of BCAA regulation initiate a diverse set of molecular mechanisms that connect changes in BCAA homeostasis to the pathogenesis of cardiovascular diseases, including myocardial infarction, ischaemia-reperfusion injury, atherosclerosis, hypertension and heart failure. In this Review, we outline the current understanding of the major factors regulating BCAA abundance and metabolic fate, highlight molecular mechanisms connecting impaired BCAA homeostasis to cardiovascular disease, discuss the epidemiological evidence connecting BCAAs with various cardiovascular disease states and identify current knowledge gaps requiring further investigation.
Collapse
Affiliation(s)
- Robert W McGarrah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA.
- Department of Medicine, Division of Cardiology, Duke University, Durham, NC, USA.
| | - Phillip J White
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
17
|
Ogawa T, Kouzu H, Osanami A, Tatekoshi Y, Sato T, Kuno A, Fujita Y, Ino S, Shimizu M, Toda Y, Ohwada W, Yano T, Tanno M, Miki T, Miura T. Downregulation of extramitochondrial BCKDH and its uncoupling from AMP deaminase in type 2 diabetic OLETF rat hearts. Physiol Rep 2023; 11:e15608. [PMID: 36802195 PMCID: PMC9938007 DOI: 10.14814/phy2.15608] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 02/20/2023] Open
Abstract
Systemic branched-chain amino acid (BCAA) metabolism is dysregulated in cardiometabolic diseases. We previously demonstrated that upregulated AMP deaminase 3 (AMPD3) impairs cardiac energetics in a rat model of obese type 2 diabetes, Otsuka Long-Evans-Tokushima fatty (OLETF). Here, we hypothesized that the cardiac BCAA levels and the activity of branched-chain α-keto acid dehydrogenase (BCKDH), a rate-limiting enzyme in BCAA metabolism, are altered by type 2 diabetes (T2DM), and that upregulated AMPD3 expression is involved in the alteration. Performing proteomic analysis combined with immunoblotting, we discovered that BCKDH localizes not only to mitochondria but also to the endoplasmic reticulum (ER), where it interacts with AMPD3. Knocking down AMPD3 in neonatal rat cardiomyocytes (NRCMs) increased BCKDH activity, suggesting that AMPD3 negatively regulates BCKDH. Compared with control rats (Long-Evans Tokushima Otsuka [LETO] rats), OLETF rats exhibited 49% higher cardiac BCAA levels and 49% lower BCKDH activity. In the cardiac ER of the OLETF rats, BCKDH-E1α subunit expression was downregulated, while AMPD3 expression was upregulated, resulting in an 80% lower AMPD3-E1α interaction compared to LETO rats. Knocking down E1α expression in NRCMs upregulated AMPD3 expression and recapitulated the imbalanced AMPD3-BCKDH expressions observed in OLETF rat hearts. E1α knockdown in NRCMs inhibited glucose oxidation in response to insulin, palmitate oxidation, and lipid droplet biogenesis under oleate loading. Collectively, these data revealed previously unrecognized extramitochondrial localization of BCKDH in the heart and its reciprocal regulation with AMPD3 and imbalanced AMPD3-BCKDH interactions in OLETF. Downregulation of BCKDH in cardiomyocytes induced profound metabolic changes that are observed in OLETF hearts, providing insight into mechanisms contributing to the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Toshifumi Ogawa
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Hidemichi Kouzu
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Arata Osanami
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Yuki Tatekoshi
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Tatsuya Sato
- Department of Cellular Physiology and Signal TransductionSapporo Medical University School of MedicineSapporoJapan
| | - Atsushi Kuno
- Department of PharmacologySapporo Medical University School of MedicineSapporoJapan
| | - Yugo Fujita
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Shoya Ino
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Masaki Shimizu
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Yuki Toda
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Wataru Ohwada
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Masaya Tanno
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Takayuki Miki
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic MedicineSapporo Medical University School of MedicineSapporoJapan
- Department of Clinical Pharmacology, Faculty of Pharmaceutical SciencesHokkaido University of ScienceSapporoJapan
| |
Collapse
|
18
|
Gaar-Humphreys KR, van den Brink A, Wekking M, Asselbergs FW, van Steenbeek FG, Harakalova M, Pei J. Targeting lipid metabolism as a new therapeutic strategy for inherited cardiomyopathies. Front Cardiovasc Med 2023; 10:1114459. [PMID: 36760574 PMCID: PMC9907444 DOI: 10.3389/fcvm.2023.1114459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Inherited cardiomyopathies caused by pathological genetic variants include multiple subtypes of heart disease. Advances in next-generation sequencing (NGS) techniques have allowed for the identification of numerous genetic variants as pathological variants. However, the disease penetrance varies among mutated genes. Some can be associated with more than one disease subtype, leading to a complex genotype-phenotype relationship in inherited cardiomyopathies. Previous studies have demonstrated disrupted metabolism in inherited cardiomyopathies and the importance of metabolic adaptations in disease onset and progression. In addition, genotype- and phenotype-specific metabolic alterations, especially in lipid metabolism, have been revealed. In this mini-review, we describe the metabolic changes that are associated with dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), which account for the largest proportion of inherited cardiomyopathies. We also summarize the affected expression of genes involved in fatty acid oxidation (FAO) in DCM and HCM, highlighting the potential of PPARA-targeting drugs as FAO modulators in treating patients with inherited cardiomyopathies.
Collapse
Affiliation(s)
- Karen R. Gaar-Humphreys
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | - Alyssa van den Brink
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mark Wekking
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | - Folkert W. Asselbergs
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Health Data Research United Kingdom and Institute of Health Informatics, University College London, London, United Kingdom
| | - Frank G. van Steenbeek
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Magdalena Harakalova
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
| | - Jiayi Pei
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
| |
Collapse
|
19
|
Fan L, Sweet DR, Fan EK, Prosdocimo DA, Madera A, Jiang Z, Padmanabhan R, Haldar SM, Vinayachandran V, Jain MK. Transcription factors KLF15 and PPARδ cooperatively orchestrate genome-wide regulation of lipid metabolism in skeletal muscle. J Biol Chem 2022; 298:101926. [PMID: 35413288 PMCID: PMC9190004 DOI: 10.1016/j.jbc.2022.101926] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle dynamically regulates systemic nutrient homeostasis through transcriptional adaptations to physiological cues. In response to changes in the metabolic environment (e.g., alterations in circulating glucose or lipid levels), networks of transcription factors and coregulators are recruited to specific genomic loci to fine-tune homeostatic gene regulation. Elucidating these mechanisms is of particular interest as these gene regulatory pathways can serve as potential targets to treat metabolic disease. The zinc-finger transcription factor Krüppel-like factor 15 (KLF15) is a critical regulator of metabolic homeostasis; however, its genome-wide distribution in skeletal muscle has not been previously identified. Here, we characterize the KLF15 cistrome in vivo in skeletal muscle and find that the majority of KLF15 binding is localized to distal intergenic regions and associated with genes related to circadian rhythmicity and lipid metabolism. We also identify critical interdependence between KLF15 and the nuclear receptor PPARδ in the regulation of lipid metabolic gene programs. We further demonstrate that KLF15 and PPARδ colocalize genome-wide, physically interact, and are dependent on one another to exert their transcriptional effects on target genes. These findings reveal that skeletal muscle KLF15 plays a critical role in metabolic adaptation through its direct actions on target genes and interactions with other nodal transcription factors such as PPARδ.
Collapse
Affiliation(s)
- Liyan Fan
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - David R Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Erica K Fan
- University of Pittsburgh School of Medicine, Department of Physical Medicine and Rehabilitation, Pittsburgh, Pennsylvania, USA
| | - Domenick A Prosdocimo
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; The Webb Law Firm, Pittsburgh, Pennsylvania, USA
| | - Annmarie Madera
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Zhen Jiang
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Roshan Padmanabhan
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Saptarsi M Haldar
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA; Department of Medicine, Division of Cardiology, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Vinesh Vinayachandran
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.
| |
Collapse
|
20
|
Joseph LC, Shi J, Nguyen QN, Pensiero V, Goulbourne C, Bauer RC, Zhang H, Morrow JP. Combined metabolomic and transcriptomic profiling approaches reveal the cardiac response to high-fat diet. iScience 2022; 25:104184. [PMID: 35494220 PMCID: PMC9038541 DOI: 10.1016/j.isci.2022.104184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/04/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022] Open
Abstract
The response of vital organs to different types of nutrition or diet is a fundamental question in physiology. We examined the cardiac response to 4 weeks of high-fat diet in mice, measuring cardiac metabolites and mRNA. Metabolomics showed dramatic differences after a high-fat diet, including increases in several acyl-carnitine species. The RNA-seq data showed changes consistent with adaptations to use more fatty acid as substrate and an increase in the antioxidant protein catalase. Changes in mRNA were correlated with changes in protein level for several highly responsive genes. We also found significant sex differences in both metabolomics and RNA-seq datasets, both at baseline and after high fat diet. This work reveals the response of a vital organ to dietary intervention at both metabolomic and transcriptomic levels, which is a fundamental question in physiology. This work also reveals significant sex differences in cardiac metabolites and gene expression.
Collapse
Affiliation(s)
- Leroy C. Joseph
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Jianting Shi
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
- Cardiometabolic Genomics Program, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | - Quynh N. Nguyen
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Victoria Pensiero
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Chris Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, USA
| | - Robert C. Bauer
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Hanrui Zhang
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
- Cardiometabolic Genomics Program, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | - John P. Morrow
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| |
Collapse
|
21
|
Jiang Z, Elsarrag SZ, Duan Q, LaGory EL, Wang Z, Alexanian M, McMahon S, Rulifson IC, Winchester S, Wang Y, Vaisse C, Brown JD, Quattrocelli M, Lin CY, Haldar SM. KLF15 cistromes reveal a hepatocyte pathway governing plasma corticosteroid transport and systemic inflammation. SCIENCE ADVANCES 2022; 8:eabj2917. [PMID: 35263131 PMCID: PMC8906731 DOI: 10.1126/sciadv.abj2917] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 01/13/2022] [Indexed: 05/15/2023]
Abstract
Circulating corticosteroids orchestrate stress adaptation, including inhibition of inflammation. While pathways governing corticosteroid biosynthesis and intracellular signaling are well understood, less is known about mechanisms controlling plasma corticosteroid transport. Here, we show that hepatocyte KLF15 (Kruppel-like factor 15) controls plasma corticosteroid transport and inflammatory responses through direct transcriptional activation of Serpina6, which encodes corticosteroid-binding globulin (CBG). Klf15-deficient mice have profoundly low CBG, reduced plasma corticosteroid binding capacity, and heightened mortality during inflammatory stress. These defects are completely rescued by reconstituting CBG, supporting that KLF15 works primarily through CBG to control plasma corticosterone homeostasis. To understand transcriptional mechanisms, we generated the first KLF15 cistromes using newly engineered Klf153xFLAG mice. Unexpectedly, liver KLF15 is predominantly promoter enriched, including Serpina6, where it binds a palindromic GC-rich motif, opens chromatin, and transactivates genes with minimal associated direct gene repression. Overall, we provide critical mechanistic insight into KLF15 function and identify a hepatocyte-intrinsic transcriptional module that potently regulates systemic corticosteroid transport and inflammation.
Collapse
Affiliation(s)
- Zhen Jiang
- Amgen Research, South San Francisco, CA 94080, USA
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Selma Z. Elsarrag
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Medical Scientist Training Program and Quantitative and Computational Biosciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qiming Duan
- Gladstone Institutes, San Francisco, CA 94158, USA
| | | | - Zhe Wang
- Amgen Research, South San Francisco, CA 94080, USA
| | | | - Sarah McMahon
- Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, UCSF School of Medicine, San Francisco, CA 94143, USA
| | | | | | - Yi Wang
- UCSF Diabetes Center and Department of Medicine, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Christian Vaisse
- UCSF Diabetes Center and Department of Medicine, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Jonathan D. Brown
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology Division, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Charles Y. Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Kronos Bio Inc., Cambridge, MA 02142, USA
| | - Saptarsi M. Haldar
- Amgen Research, South San Francisco, CA 94080, USA
- Gladstone Institutes, San Francisco, CA 94158, USA
- Cardiology Division, Department of Medicine, UCSF School of Medicine, San Francisco, CA 94143, USA
| |
Collapse
|
22
|
Palioura D, Lazou A, Drosatos K. Krüppel-like factor (KLF)5: An emerging foe of cardiovascular health. J Mol Cell Cardiol 2022; 163:56-66. [PMID: 34653523 PMCID: PMC8816822 DOI: 10.1016/j.yjmcc.2021.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/22/2021] [Accepted: 10/07/2021] [Indexed: 02/03/2023]
Abstract
Krüppel-like factors (KLFs) are DNA-binding transcriptional factors, which regulate various pathways that pertain to development, metabolism and other cellular mechanisms. KLF5 was first cloned in 1993 and by 1999, it was reported as the intestinal-enriched KLF. Beyond findings that have associated KLF5 with normal development and cancer, it has been associated with various types of cardiovascular (CV) complications and regulation of metabolic pathways in the liver, heart, adipose tissue and skeletal muscle. Specifically, increased KLF5 expression has been linked with cardiomyopathy in diabetes, end-stage heart failure, and as well as in vascular atherosclerotic lesions. In this review article, we summarize research findings about transcriptional, post-transcriptional and post-translational regulation of KLF5, as well as the role of KLF5 in the biology of cells and organs that affect cardiovascular health either directly or indirectly. Finally, we propose KLF5 inhibition as an emerging approach for cardiovascular therapeutics.
Collapse
Affiliation(s)
- Dimitra Palioura
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA;,School of Biology, Aristotle University of Thessaloniki, GR, Greece
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, GR, Greece
| | - Konstantinos Drosatos
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
23
|
Yamamoto T, Sano M. Deranged Myocardial Fatty Acid Metabolism in Heart Failure. Int J Mol Sci 2022; 23:996. [PMID: 35055179 PMCID: PMC8779056 DOI: 10.3390/ijms23020996] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
The heart requires fatty acids to maintain its activity. Various mechanisms regulate myocardial fatty acid metabolism, such as energy production using fatty acids as fuel, for which it is known that coordinated control of fatty acid uptake, β-oxidation, and mitochondrial oxidative phosphorylation steps are important for efficient adenosine triphosphate (ATP) production without unwanted side effects. The fatty acids taken up by cardiomyocytes are not only used as substrates for energy production but also for the synthesis of triglycerides and the replacement reaction of fatty acid chains in cell membrane phospholipids. Alterations in fatty acid metabolism affect the structure and function of the heart. Recently, breakthrough studies have focused on the key transcription factors that regulate fatty acid metabolism in cardiomyocytes and the signaling systems that modify their functions. In this article, we reviewed the latest research on the role of fatty acid metabolism in the pathogenesis of heart failure and provide an outlook on future challenges.
Collapse
Affiliation(s)
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| |
Collapse
|
24
|
Piret SE, Attallah AA, Gu X, Guo Y, Gujarati NA, Henein J, Zollman A, Hato T, Ma'ayan A, Revelo MP, Dickman KG, Chen CH, Shun CT, Rosenquist TA, He JC, Mallipattu SK. Loss of proximal tubular transcription factor Krüppel-like factor 15 exacerbates kidney injury through loss of fatty acid oxidation. Kidney Int 2021; 100:1250-1267. [PMID: 34634362 PMCID: PMC8608748 DOI: 10.1016/j.kint.2021.08.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/06/2021] [Accepted: 08/20/2021] [Indexed: 11/24/2022]
Abstract
Loss of fatty acid β-oxidation (FAO) in the proximal tubule is a critical mediator of acute kidney injury and eventual fibrosis. However, transcriptional mediators of FAO in proximal tubule injury remain understudied. Krüppel-like factor 15 (KLF15), a highly enriched zinc-finger transcription factor in the proximal tubule, was significantly reduced in proximal tubule cells after aristolochic acid I (AAI) treatment, a proximal tubule-specific injury model. Proximal tubule specific knockout of Klf15 exacerbated proximal tubule injury and kidney function decline compared to control mice during the active phase of AAI treatment, and after ischemia-reperfusion injury. Furthermore, along with worsening proximal tubule injury and kidney function decline, knockout mice exhibited increased kidney fibrosis as compared to control mice during the remodeling phase after AAI treatment. RNA-sequencing of kidney cortex demonstrated increased transcripts involved in immune system and integrin signaling pathways and decreased transcripts encompassing metabolic pathways, specifically FAO, and PPARα signaling, in knockout versus control mice after AAI treatment. In silico and experimental chromatin immunoprecipitation studies collectively demonstrated that KLF15 occupied the promoter region of key FAO genes, CPT1A and ACAA2, in close proximity to transcription factor PPARα binding sites. While the loss of Klf15 reduced the expression of Cpt1a and Acaa2 and led to compromised FAO, induction of KLF15 partially rescued loss of FAO in AAI-treated cells. Klf15, Ppara, Cpt1a, and Acaa2 expression was also decreased in other mouse kidney injury models. Tubulointerstitial KLF15 independently correlated with eGFR, PPARA and CPT1A appearance in expression arrays from human kidney biopsies. Thus, proximal tubule-specific loss of Klf15 exacerbates acute kidney injury and fibrosis, likely due to loss of interaction with PPARα leading to loss of FAO gene transcription.
Collapse
Affiliation(s)
- Sian E Piret
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Ahmed A Attallah
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Xiangchen Gu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, USA; Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Yiqing Guo
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Nehaben A Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Justina Henein
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Amy Zollman
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Takashi Hato
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Kathleen G Dickman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | - Chung-Hsin Chen
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Tung Shun
- Department of Forensic Medicine and Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Thomas A Rosenquist
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | - John C He
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, USA; Renal Division, Northport VA Medical Center, Northport, New York, USA.
| |
Collapse
|
25
|
Sun N, Shen C, Zhang L, Wu X, Yu Y, Yang X, Yang C, Zhong C, Gao Z, Miao W, Yang Z, Gao W, Hu L, Williams K, Liu C, Chang Y, Gao Y. Hepatic Krüppel-like factor 16 (KLF16) targets PPARα to improve steatohepatitis and insulin resistance. Gut 2021; 70:2183-2195. [PMID: 33257471 PMCID: PMC8515101 DOI: 10.1136/gutjnl-2020-321774] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 10/20/2020] [Accepted: 11/08/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Impaired hepatic fatty acids oxidation results in lipid accumulation and redox imbalance, promoting the development of fatty liver diseases and insulin resistance. However, the underlying pathogenic mechanism is poorly understood. Krüppel-like factor 16 (KLF16) is a transcription factor that abounds in liver. We explored whether and by what mechanisms KLF16 affects hepatic lipid catabolism to improve hepatosteatosis and insulin resistance. DESIGN KLF16 expression was determined in patients with non-alcoholic fatty liver disease (NAFLD) and mice models. The role of KLF16 in the regulation of lipid metabolism was investigated using hepatocyte-specific KLF16-deficient mice fed a high-fat diet (HFD) or using an adenovirus/adeno-associated virus to alter KLF16 expression in mouse primary hepatocytes (MPHs) and in vivo livers. RNA-seq, luciferase reporter gene assay and ChIP analysis served to explore the molecular mechanisms involved. RESULTS KLF16 expression was decreased in patients with NAFLD, mice models and oleic acid and palmitic acid (OA and PA) cochallenged hepatocytes. Hepatic KLF16 knockout impaired fatty acid oxidation, aggravated mitochondrial stress, ROS burden, advancing hepatic steatosis and insulin resistance. Conversely, KLF16 overexpression reduced lipid deposition and improved insulin resistance via directly binding the promoter of peroxisome proliferator-activated receptor α (PPARα) to accelerate fatty acids oxidation and attenuate mitochondrial stress, oxidative stress in db/db and HFD mice. PPARα deficiency diminished the KLF16-evoked protective effects against lipid deposition in MPHs. Hepatic-specific PPARα overexpression effectively rescued KLF16 deficiency-induced hepatic steatosis, altered redox balance and insulin resistance. CONCLUSIONS These findings prove that a direct KLF16-PPARα pathway closely links hepatic lipid homeostasis and redox balance, whose dysfunction promotes insulin resistance and hepatic steatosis.
Collapse
Affiliation(s)
- Nannan Sun
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuangpeng Shen
- Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Zhang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xiaojie Wu
- Central Lab of Binzhou People’s Hospital, Central Lab of Binzhou People’s Hospital, Shandong, China
| | - Yuanyuan Yu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chen Yang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chong Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhao Gao
- Guangdong Provincial Institute of Sports Science, Guangzhou, Guangdong, China
| | - Wei Miao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zehong Yang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weihang Gao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kevin Williams
- Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Changhui Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yongsheng Chang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yong Gao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China .,Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
26
|
Supruniuk E, Żebrowska E, Chabowski A. Branched chain amino acids-friend or foe in the control of energy substrate turnover and insulin sensitivity? Crit Rev Food Sci Nutr 2021; 63:2559-2597. [PMID: 34542351 DOI: 10.1080/10408398.2021.1977910] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Branched chain amino acids (BCAA) and their derivatives are bioactive molecules with pleiotropic functions in the human body. Elevated fasting blood BCAA concentrations are considered as a metabolic hallmark of obesity, insulin resistance, dyslipidaemia, nonalcoholic fatty liver disease, type 2 diabetes and cardiovascular disease. However, since increased BCAA amount is observed both in metabolically healthy and obese subjects, a question whether BCAA are mechanistic drivers of insulin resistance and its morbidities or only markers of metabolic dysregulation, still remains open. The beneficial effects of BCAA on body weight and composition, aerobic capacity, insulin secretion and sensitivity demand high catabolic potential toward amino acids and/or adequate BCAA intake. On the opposite, BCAA-related inhibition of lipogenesis and lipolysis enhancement may preclude impairment in insulin sensitivity. Thereby, the following review addresses various strategies pertaining to the modulation of BCAA catabolism and the possible roles of BCAA in energy homeostasis. We also aim to elucidate mechanisms behind the heterogeneity of ramifications associated with BCAA modulation.
Collapse
Affiliation(s)
- Elżbieta Supruniuk
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Żebrowska
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
27
|
Li L, Xu W, Zhang L. KLF15 Regulates Oxidative Stress Response in Cardiomyocytes through NAD . Metabolites 2021; 11:metabo11090620. [PMID: 34564436 PMCID: PMC8468172 DOI: 10.3390/metabo11090620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
KLF15 has recently emerged as a central regulator of metabolism. Although its connection to oxidative stress has been suspected, there has not been any study to date that directly demonstrates the molecular link. In this study, we sought to determine the role of KLF15 in cardiac oxidative stress. We found that KLF15 deficiency in the heart is associated with increased oxidative stress. Acute deficiency of KLF15 in neonatal rat ventricular myocytes (NRVMs) leads to the defective clearance of reactive oxygen species (ROS) and an exaggerated cell death following a variety of oxidative stresses. Mechanistically, we found that KLF15 deficiency leads to reduced amounts of the rate-limiting NAD+ salvage enzyme NAMPT and to NAD+ deficiency. The resultant SIRT3-dependent hyperacetylation and the inactivation of mitochondrial antioxidants can be rescued by MnSOD mimetics or NAD+ precursors. Collectively, these findings suggest that KLF15 regulates cardiac ROS clearance through the regulation of NAD+ levels. Our findings establish KLF15 as a central coordinator of cardiac metabolism and ROS clearance.
Collapse
|
28
|
Abstract
Mammalian cardiomyocytes mostly utilize oxidation of fatty acids to generate ATP. The fetal heart, in stark contrast, mostly uses anaerobic glycolysis. During perinatal development, thyroid hormone drives extensive metabolic remodeling in the heart for adaptation to extrauterine life. These changes coincide with critical functional maturation and exit of the cell cycle, making the heart a post-mitotic organ. Here, we review the current understanding on the perinatal shift in metabolism, hormonal status, and proliferative potential in cardiomyocytes. Thyroid hormone and glucocorticoids have roles in adult cardiac metabolism, and both pathways have been implicated as regulators of myocardial regeneration. We discuss the evidence that suggests these processes could be interrelated and how this can help explain variation in cardiac regeneration across ontogeny and phylogeny, and we note what breakthroughs are still to be made.
Collapse
Affiliation(s)
- Niall Graham
- Cardiovascular Research Institute and Department of Physiology, University of California San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Correspondence: Guo N Huang, Ph.D., University of California San Francisco, 555 Mission Bay Blvd South, Room 352V, San Francisco, CA 94158, USA.
| |
Collapse
|
29
|
Chow LS, Bosnakovski D, Mashek DG, Kyba M, Perlingeiro RCR, Magli A. Chromatin accessibility profiling identifies evolutionary conserved loci in activated human satellite cells. Stem Cell Res 2021; 55:102496. [PMID: 34411972 PMCID: PMC8917817 DOI: 10.1016/j.scr.2021.102496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/19/2021] [Accepted: 08/04/2021] [Indexed: 12/04/2022] Open
Abstract
Satellite cells represent the main myogenic population accounting for skeletal muscle homeostasis and regeneration. While our knowledge of the signaling pathways controlling satellite cell regenerative capability is increasing, the underlying epigenetic mechanisms are still not clear, especially in the case of human satellite cells. Here, by performing chromatin accessibility profiling (ATAC-seq) in samples isolated from human and murine muscles, we investigated the changes in the epigenetic landscape occurring during the transition from activated satellite cells to myoblasts. Our analysis identifies a compendium of putative regulatory elements defining human activated satellite cells and myoblasts, respectively. A subset of these differentially accessible loci is shared by both murine and human satellite cells, includes elements associated with known self-renewal regulators, and is enriched for motifs bound by transcription factors participating in satellite cell regulation. Integration of transcriptional and epigenetic data reveals that known regulators of metabolic gene expression, such as PPARGC1A, represent potential PAX7 targets. Through characterization of genomic networks and the underlying effectors, our data represent an important starting point for decoding and manipulating the molecular mechanisms underlying human satellite cell muscle regenerative potential.
Collapse
Affiliation(s)
- Lisa S Chow
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Darko Bosnakovski
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA; University Goce Delcev - Shtip, Faculty of Medical Sciences, Shtip, Macedonia
| | - Douglas G Mashek
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Alessandro Magli
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
30
|
Abstract
Circadian rhythm evolved to allow organisms to coordinate intrinsic physiological functions in anticipation of recurring environmental changes. The importance of this coordination is exemplified by the tight temporal control of cardiac metabolism. Levels of metabolites, metabolic flux, and response to nutrients all oscillate in a time-of-day-dependent fashion. While these rhythms are affected by oscillatory behavior (feeding/fasting, wake/sleep) and neurohormonal changes, recent data have unequivocally demonstrated an intrinsic circadian regulation at the tissue and cellular level. The circadian clock - through a network of a core clock, slave clock, and effectors - exerts intricate temporal control of cardiac metabolism, which is also integrated with environmental cues. The combined anticipation and adaptability that the circadian clock enables provide maximum advantage to cardiac function. Disruption of the circadian rhythm, or dyssynchrony, leads to cardiometabolic disorders seen not only in shift workers but in most individuals in modern society. In this Review, we describe current findings on rhythmic cardiac metabolism and discuss the intricate regulation of circadian rhythm and the consequences of rhythm disruption. An in-depth understanding of the circadian biology in cardiac metabolism is critical in translating preclinical findings from nocturnal-animal models as well as in developing novel chronotherapeutic strategies.
Collapse
Affiliation(s)
- Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Department of Medicine.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, and.,School of Medicine; Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
31
|
Xiao C, Wei T, Liu LX, Liu JQ, Wang CX, Yuan ZY, Ma HH, Jin HG, Zhang LC, Cao Y. Whole-Transcriptome Analysis of Preadipocyte and Adipocyte and Construction of Regulatory Networks to Investigate Lipid Metabolism in Sheep. Front Genet 2021; 12:662143. [PMID: 34394181 PMCID: PMC8358208 DOI: 10.3389/fgene.2021.662143] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/09/2021] [Indexed: 01/20/2023] Open
Abstract
Many local sheep breeds in China have poor meat quality. Increasing intramuscular fat (IMF) content can significantly improve the quality of mutton. However, the molecular mechanisms of intramuscular adipocyte formation and differentiation remain unclear. This study compared differences between preadipocytes and mature adipocytes by whole-transcriptome sequencing and constructed systematically regulatory networks according to the relationship predicted among the differentially expressed RNAs (DERs). Sequencing results showed that in this process, there were 1,196, 754, 100, and 17 differentially expressed messenger RNAs (mRNAs), long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), respectively. Gene Ontology analysis showed that most DERs enriched in Cell Part, Cellular Process, Biological Regulation, and Binding terms. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis found that the DERs primarily focused on Focal adhesion, phosphoinositide 3-kinase (PI3K)-Akt, mitogen-activated protein kinase (MAPK), peroxisome proliferator-activated receptor (PPAR) signaling pathways. Forty (40) DERs were randomly selected from the core regulatory network to verify the accuracy of the sequence data. The results of qPCR showed that the DER expression trend was consistent with sequence data. Four novel promising candidate miRNAs (miR-336, miR-422, miR-578, and miR-722) played crucial roles in adipocyte differentiation, and they also participated in multiple and important regulatory networks. We verified the expression pattern of the miRNAs and related pathways’ members at five time points in the adipocyte differentiation process (0, 2, 4, 6, 8, 10 days) by qPCR, including miR-336/ACSL4/LncRNA-MSTRG71379/circRNA0002331, miR-422/FOXO4/LncRNA-MSTRG54995/circRNA0000520, miR-578/IGF1/LncRNA-MSTRG102235/circRNA0002971, and miR-722/PDK4/LncRNA-MSTRG107440/circ RNA0002909. In this study, our data provided plenty of valuable candidate DERs and regulatory networks for researching the molecular mechanisms of sheep adipocyte differentiation and will assist studies in improving the IMF.
Collapse
Affiliation(s)
- Cheng Xiao
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Tian Wei
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Li Xiang Liu
- Jilin Academy of Agricultural Sciences, Gongzhuling, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jian Qiang Liu
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Chun Xin Wang
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Zhi Yu Yuan
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Hui Hai Ma
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Hai Guo Jin
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Li Chun Zhang
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Yang Cao
- Jilin Academy of Agricultural Sciences, Gongzhuling, China
| |
Collapse
|
32
|
Nabatame Y, Hosooka T, Aoki C, Hosokawa Y, Imamori M, Tamori Y, Okamatsu‐Ogura Y, Yoneshiro T, Kajimura S, Saito M, Ogawa W. Kruppel-like factor 15 regulates fuel switching between glucose and fatty acids in brown adipocytes. J Diabetes Investig 2021; 12:1144-1151. [PMID: 33480176 PMCID: PMC8264414 DOI: 10.1111/jdi.13511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/04/2020] [Accepted: 01/07/2021] [Indexed: 01/16/2023] Open
Abstract
AIMS/INTRODUCTION Brown adipose tissue (BAT) utilizes large amounts of fuel for thermogenesis, but the mechanism by which fuel substrates are switched in response to changes in energy status is poorly understood. We have now investigated the role of Kruppel-like factor 15 (KLF15), a transcription factor expressed at a high level in adipose tissue, in the regulation of fuel utilization in BAT. MATERIALS AND METHODS Depletion or overexpression of KLF15 in HB2 differentiated brown adipocytes was achieved by adenoviral infection. Glucose and fatty acid oxidation were measured with radioactive substrates, pyruvate dehydrogenase complex activity was determined with a colorimetric assay, and gene expression was examined by reverse transcription and real-time polymerase chain reaction analysis. RESULTS Knockdown of KLF15 in HB2 cells attenuated fatty acid oxidation in association with downregulation of the expression of genes related to this process including Acox1 and Fatp1, whereas it increased glucose oxidation. Expression of the gene for pyruvate dehydrogenase kinase 4 (PDK4), a negative regulator of pyruvate dehydrogenase complex, was increased or decreased by KLF15 overexpression or knockdown, respectively, in HB2 cells, with these changes being accompanied by a respective decrease or increase in pyruvate dehydrogenase complex activity. Chromatin immunoprecipitation showed that Pdk4 is a direct target of KLF15 in HB2 cells. Finally, fasting increased expression of KLf15, Pdk4 and genes involved in fatty acid utilization in BAT of mice, whereas refeeding suppressed Klf15 and Pdk4 expression. CONCLUSIONS Our results implicate KLF15 in the regulation of fuel switching between glucose and fatty acids in response to changes in energy status in BAT.
Collapse
Affiliation(s)
- Yuko Nabatame
- Division of Diabetes and EndocrinologyKobe University Graduate School of MedicineKobeJapan
| | - Tetsuya Hosooka
- Division of Diabetes and EndocrinologyKobe University Graduate School of MedicineKobeJapan
- Division of Development of Advanced Therapy for Metabolic DiseaseKobe University Graduate School of MedicineKobeJapan
| | - Chikako Aoki
- Division of Diabetes and EndocrinologyKobe University Graduate School of MedicineKobeJapan
| | - Yusei Hosokawa
- Division of Diabetes and EndocrinologyKobe University Graduate School of MedicineKobeJapan
| | - Makoto Imamori
- Division of Diabetes and EndocrinologyKobe University Graduate School of MedicineKobeJapan
| | - Yoshikazu Tamori
- Division of Diabetes and EndocrinologyKobe University Graduate School of MedicineKobeJapan
- Division of Creative Health PromotionKobe University Graduate School of MedicineKobeJapan
| | - Yuko Okamatsu‐Ogura
- Laboratory of BiochemistryFaculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Takeshi Yoneshiro
- UCSF Diabetes CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Cell and Tissue BiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Present address:
Division of Metabolic MedicineResearch Center for Advanced Science and TechnologyThe University of TokyoTokyoJapan
| | - Shingo Kajimura
- UCSF Diabetes CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Cell and Tissue BiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Present address:
Division of Endocrinology, Diabetes and MetabolismBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Masayuki Saito
- Laboratory of BiochemistryFaculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Wataru Ogawa
- Division of Diabetes and EndocrinologyKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
33
|
Gillard GB, Grønvold L, Røsæg LL, Holen MM, Monsen Ø, Koop BF, Rondeau EB, Gundappa MK, Mendoza J, Macqueen DJ, Rohlfs RV, Sandve SR, Hvidsten TR. Comparative regulomics supports pervasive selection on gene dosage following whole genome duplication. Genome Biol 2021; 22:103. [PMID: 33849620 PMCID: PMC8042706 DOI: 10.1186/s13059-021-02323-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Whole genome duplication (WGD) events have played a major role in eukaryotic genome evolution, but the consequence of these extreme events in adaptive genome evolution is still not well understood. To address this knowledge gap, we used a comparative phylogenetic model and transcriptomic data from seven species to infer selection on gene expression in duplicated genes (ohnologs) following the salmonid WGD 80-100 million years ago. RESULTS We find rare cases of tissue-specific expression evolution but pervasive expression evolution affecting many tissues, reflecting strong selection on maintenance of genome stability following genome doubling. Ohnolog expression levels have evolved mostly asymmetrically, by diverting one ohnolog copy down a path towards lower expression and possible pseudogenization. Loss of expression in one ohnolog is significantly associated with transposable element insertions in promoters and likely driven by selection on gene dosage including selection on stoichiometric balance. We also find symmetric expression shifts, and these are associated with genes under strong evolutionary constraints such as ribosome subunit genes. This possibly reflects selection operating to achieve a gene dose reduction while avoiding accumulation of "toxic mutations". Mechanistically, ohnolog regulatory divergence is dictated by the number of bound transcription factors in promoters, with transposable elements being one likely source of novel binding sites driving tissue-specific gains in expression. CONCLUSIONS Our results imply pervasive adaptive expression evolution following WGD to overcome the immediate challenges posed by genome doubling and to exploit the long-term genetic opportunities for novel phenotype evolution.
Collapse
Affiliation(s)
- Gareth B. Gillard
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Lars Grønvold
- Center for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Line L. Røsæg
- Center for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Matilde Mengkrog Holen
- Center for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Øystein Monsen
- Center for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Ben F. Koop
- Department of Biology, University of Victoria, Victoria, Canada
| | - Eric B. Rondeau
- Department of Biology, University of Victoria, Victoria, Canada
| | - Manu Kumar Gundappa
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - John Mendoza
- Department of Computer Science, San Francisco State University, San Francisco, USA
| | - Daniel J. Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Rori V. Rohlfs
- Department of Biology, San Francisco State University, San Francisco, USA
| | - Simen R. Sandve
- Center for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Torgeir R. Hvidsten
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
34
|
Pei J, Schuldt M, Nagyova E, Gu Z, El Bouhaddani S, Yiangou L, Jansen M, Calis JJA, Dorsch LM, Blok CS, van den Dungen NAM, Lansu N, Boukens BJ, Efimov IR, Michels M, Verhaar MC, de Weger R, Vink A, van Steenbeek FG, Baas AF, Davis RP, Uh HW, Kuster DWD, Cheng C, Mokry M, van der Velden J, Asselbergs FW, Harakalova M. Multi-omics integration identifies key upstream regulators of pathomechanisms in hypertrophic cardiomyopathy due to truncating MYBPC3 mutations. Clin Epigenetics 2021; 13:61. [PMID: 33757590 PMCID: PMC7989210 DOI: 10.1186/s13148-021-01043-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/28/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most common genetic disease of the cardiac muscle, frequently caused by mutations in MYBPC3. However, little is known about the upstream pathways and key regulators causing the disease. Therefore, we employed a multi-omics approach to study the pathomechanisms underlying HCM comparing patient hearts harboring MYBPC3 mutations to control hearts. RESULTS Using H3K27ac ChIP-seq and RNA-seq we obtained 9310 differentially acetylated regions and 2033 differentially expressed genes, respectively, between 13 HCM and 10 control hearts. We obtained 441 differentially expressed proteins between 11 HCM and 8 control hearts using proteomics. By integrating multi-omics datasets, we identified a set of DNA regions and genes that differentiate HCM from control hearts and 53 protein-coding genes as the major contributors. This comprehensive analysis consistently points toward altered extracellular matrix formation, muscle contraction, and metabolism. Therefore, we studied enriched transcription factor (TF) binding motifs and identified 9 motif-encoded TFs, including KLF15, ETV4, AR, CLOCK, ETS2, GATA5, MEIS1, RXRA, and ZFX. Selected candidates were examined in stem cell-derived cardiomyocytes with and without mutated MYBPC3. Furthermore, we observed an abundance of acetylation signals and transcripts derived from cardiomyocytes compared to non-myocyte populations. CONCLUSIONS By integrating histone acetylome, transcriptome, and proteome profiles, we identified major effector genes and protein networks that drive the pathological changes in HCM with mutated MYBPC3. Our work identifies 38 highly affected protein-coding genes as potential plasma HCM biomarkers and 9 TFs as potential upstream regulators of these pathomechanisms that may serve as possible therapeutic targets.
Collapse
Affiliation(s)
- J Pei
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Regenerative Medicine Utrecht (RMU), University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Department of Nephrology and Hypertension, DIG-D, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - M Schuldt
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - E Nagyova
- Laboratory of Clinical Chemistry and Hematology, UMC Utrecht, Utrecht, The Netherlands
| | - Z Gu
- Department of Biostatistics and Research Support, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - S El Bouhaddani
- Department of Biostatistics and Research Support, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - L Yiangou
- Department of Anatomy and Embryology, LUMC, Leiden, The Netherlands
| | - M Jansen
- Department of Genetics, Division of Laboratories, Pharmacy and Biomedical Genetics, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - J J A Calis
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Regenerative Medicine Utrecht (RMU), University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
| | - L M Dorsch
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - C Snijders Blok
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
| | - N A M van den Dungen
- Laboratory of Clinical Chemistry and Hematology, UMC Utrecht, Utrecht, The Netherlands
| | - N Lansu
- Laboratory of Clinical Chemistry and Hematology, UMC Utrecht, Utrecht, The Netherlands
| | - B J Boukens
- Department of Medical Biology, AMC, Amsterdam, The Netherlands
| | - I R Efimov
- Department of Biomedical Engineering, GWU, Washington, DC, USA
| | - M Michels
- Department of Cardiology, Thoraxcentre, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - M C Verhaar
- Regenerative Medicine Utrecht (RMU), University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Department of Nephrology and Hypertension, DIG-D, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - R de Weger
- Department of Pathology, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - A Vink
- Department of Pathology, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - F G van Steenbeek
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Regenerative Medicine Utrecht (RMU), University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Utrecht, Utrecht, The Netherlands
| | - A F Baas
- Department of Genetics, Division of Laboratories, Pharmacy and Biomedical Genetics, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - R P Davis
- Department of Anatomy and Embryology, LUMC, Leiden, The Netherlands
| | - H W Uh
- Department of Biostatistics and Research Support, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - D W D Kuster
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - C Cheng
- Regenerative Medicine Utrecht (RMU), University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Department of Nephrology and Hypertension, DIG-D, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
- Department of Biomedical Engineering, GWU, Washington, DC, USA
| | - M Mokry
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Regenerative Medicine Utrecht (RMU), University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Hematology, UMC Utrecht, Utrecht, The Netherlands
- Division of Paediatrics, UMC Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - J van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - F W Asselbergs
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands.
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK.
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK.
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Room E03.818, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands.
| | - M Harakalova
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands.
- Regenerative Medicine Utrecht (RMU), University Medical Center Utrecht, University of Utrecht, 3584 CT, Utrecht, The Netherlands.
| |
Collapse
|
35
|
KLF15 Loss-of-Function Mutation Underlying Atrial Fibrillation as well as Ventricular Arrhythmias and Cardiomyopathy. Genes (Basel) 2021; 12:genes12030408. [PMID: 33809104 PMCID: PMC8001991 DOI: 10.3390/genes12030408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/27/2021] [Accepted: 03/09/2021] [Indexed: 12/04/2022] Open
Abstract
Atrial fibrillation (AF) represents the most common type of clinical cardiac arrhythmia and substantially increases the risks of cerebral stroke, heart failure and death. Accumulating evidence has convincingly demonstrated the strong genetic basis of AF, and an increasing number of pathogenic variations in over 50 genes have been causally linked to AF. Nevertheless, AF is of pronounced genetic heterogeneity, and the genetic determinants underpinning AF in most patients remain obscure. In the current investigation, a Chinese pedigree with AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy was recruited. Whole exome sequencing and bioinformatic analysis of the available family members were conducted, and a novel heterozygous variation in the KLF15 gene (encoding Krüppel-like factor 15, a transcription factor critical for cardiac electrophysiology and structural remodeling), NM_014079.4: c.685A>T; p.(Lys229*), was identified. The variation was verified by Sanger sequencing and segregated with autosomal dominant AF in the family with complete penetrance. The variation was absent from 300 unrelated healthy subjects used as controls. In functional assays using a dual-luciferase assay system, mutant KLF15 showed neither transcriptional activation of the KChIP2 promoter nor transcriptional inhibition of the CTGF promoter, alone or in the presence of TGFB1, a key player in the pathogenesis of arrhythmias and cardiomyopathies. The findings indicate KLF15 as a new causative gene responsible for AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy, and they provide novel insight into the molecular mechanisms underlying cardiac arrhythmias and hypertrophic cardiomyopathy.
Collapse
|
36
|
Kretzschmar T, Wu JMF, Schulze PC. Mitochondrial Homeostasis Mediates Lipotoxicity in the Failing Myocardium. Int J Mol Sci 2021; 22:1498. [PMID: 33540894 PMCID: PMC7867320 DOI: 10.3390/ijms22031498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/17/2023] Open
Abstract
Heart failure remains the most common cause of death in the industrialized world. In spite of new therapeutic interventions that are constantly being developed, it is still not possible to completely protect against heart failure development and progression. This shows how much more research is necessary to understand the underlying mechanisms of this process. In this review, we give a detailed overview of the contribution of impaired mitochondrial dynamics and energy homeostasis during heart failure progression. In particular, we focus on the regulation of fatty acid metabolism and the effects of fatty acid accumulation on mitochondrial structural and functional homeostasis.
Collapse
Affiliation(s)
| | | | - P. Christian Schulze
- Department of Internal Medicine I, University Hospital Jena, 07747 Jena, Thüringen, Germany; (T.K.); (J.M.F.W.)
| |
Collapse
|
37
|
The role of FATP1 in lipid accumulation: a review. Mol Cell Biochem 2021; 476:1897-1903. [PMID: 33486652 DOI: 10.1007/s11010-021-04057-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
Lipid accumulation in mammals has been widely studied for decades due to its significant association with obesity in humans and meat quality in livestock animals. Fatty acid transport 1 (FATP1) is an evolutionarily conserved protein that localizes to the plasma membrane to enhance the transportation of fatty acids (FAs). In line with this function, FATP1 is involved in the metabolism of FAs, including their esterification and oxidation. In addition, the expression of FATP1 can be regulated by several energy-related factors, such as insulin and PPAR activators and transcription factors. These events connect FATP1 with cellular lipid accumulation. Recently, several studies have suggested that FATP1 acts as a facilitator in cellular lipid accumulation, whereas others hold a contrary view. Here, we will review these data and probe the possibility that FATP1 acts as a regulator in lipid accumulation, which will provide effective information for studies on the relationship between FATP1 and obesity in humans and meat quality in livestock animals.
Collapse
|
38
|
Xu W, Jain MK, Zhang L. Molecular link between circadian clocks and cardiac function: a network of core clock, slave clock, and effectors. Curr Opin Pharmacol 2020; 57:28-40. [PMID: 33189913 DOI: 10.1016/j.coph.2020.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/27/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023]
Abstract
The circadian rhythm has a strong influence on both cardiac physiology and disease in humans. Preclinical studies primarily using tissue-specific transgenic mouse models have contributed to our understanding of the molecular mechanism of the circadian clock in the cardiovascular system. The core clock driven by CLOCK:BMAL1 complex functions as a universal timing machinery that primarily sets the pace in all mammalian cell types. In one specific cell or tissue type, core clock may control a secondary transcriptional oscillator, conceptualized as slave clock, which confers the oscillatory expression of tissue-specific effectors. Here, we discuss a core clock-slave clock-effectors network, which links the molecular clock to cardiac function.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Department of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, USA; School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
39
|
Biswas D, Dao KT, Mercer A, Cowie AM, Duffley L, El Hiani Y, Kienesberger PC, Pulinilkunnil T. Branched-chain ketoacid overload inhibits insulin action in the muscle. J Biol Chem 2020; 295:15597-15621. [PMID: 32878988 DOI: 10.1074/jbc.ra120.013121] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/29/2020] [Indexed: 12/18/2022] Open
Abstract
Branched-chain α-keto acids (BCKAs) are catabolites of branched-chain amino acids (BCAAs). Intracellular BCKAs are cleared by branched-chain ketoacid dehydrogenase (BCKDH), which is sensitive to inhibitory phosphorylation by BCKD kinase (BCKDK). Accumulation of BCKAs is an indicator of defective BCAA catabolism and has been correlated with glucose intolerance and cardiac dysfunction. However, it is unclear whether BCKAs directly alter insulin signaling and function in the skeletal and cardiac muscle cell. Furthermore, the role of excess fatty acids (FAs) in perturbing BCAA catabolism and BCKA availability merits investigation. By using immunoblotting and ultra-performance liquid chromatography MS/MS to analyze the hearts of fasted mice, we observed decreased BCAA-catabolizing enzyme expression and increased circulating BCKAs, but not BCAAs. In mice subjected to diet-induced obesity (DIO), we observed similar increases in circulating BCKAs with concomitant changes in BCAA-catabolizing enzyme expression only in the skeletal muscle. Effects of DIO were recapitulated by simulating lipotoxicity in skeletal muscle cells treated with saturated FA, palmitate. Exposure of muscle cells to high concentrations of BCKAs resulted in inhibition of insulin-induced AKT phosphorylation, decreased glucose uptake, and mitochondrial oxygen consumption. Altering intracellular clearance of BCKAs by genetic modulation of BCKDK and BCKDHA expression showed similar effects on AKT phosphorylation. BCKAs increased protein translation and mTORC1 activation. Pretreating cells with mTORC1 inhibitor rapamycin restored BCKA's effect on insulin-induced AKT phosphorylation. This study provides evidence for FA-mediated regulation of BCAA-catabolizing enzymes and BCKA content and highlights the biological role of BCKAs in regulating muscle insulin signaling and function.
Collapse
Affiliation(s)
- Dipsikha Biswas
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Khoi T Dao
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Angella Mercer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Andrew M Cowie
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Luke Duffley
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Yassine El Hiani
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada.
| |
Collapse
|
40
|
Li H, Pan Y, Bao L, Li Y, Cheng C, Liu L, Xiang J, Cheng J, Zhang J, Chu W, Shen Y. Impact of short-term starvation and refeeding on the expression of KLF15 and regulatory mechanism of branched-chain amino acids metabolism in muscle of Chinese soft-shelled turtle (Pelodiscus sinensis). Gene 2020; 752:144782. [PMID: 32442577 DOI: 10.1016/j.gene.2020.144782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/21/2020] [Accepted: 05/14/2020] [Indexed: 10/25/2022]
Abstract
The branched-chain amino acids (BCAA) play an important role in muscle energy metabolism, and Krüppel-like factor 15 (KLF15) is an essential regulator of BCAA metabolism in muscle under nutritional deficiency. In this study, we analyzed the effect of normal feeding (starvation for 0 day), starvation for 3, 7, 10, 15 days, and refeeding for 7 days after 15 days of starvation on the expression of KLF15 and BCAA metabolism in muscle of Chinese soft-shelled turtles by a fasting-refeeding trial. The results showed that the level of KLF15 transcription was increased first and then decreased in muscle during short-term starvation, and the protein level was gradually increased. Both the mRNA and protein level of the KLF15 returned to normal feeding level after refeeding for 7 days. The changing trend of the activities of branched-chain aminotransferase (BCAT) and alanine aminotransferase (ALT) was consistent to that of KLF15 mRNA, but at the transcription level, the expression of BCAT mRNA was consistent with the change of enzyme activity as well as ALT continued to increase in muscle under starvation. In addition, BCAA content showed a trend that decreased first and then increased under starvation, while the alanine (Ala) was the contrary. The above results indicated that the regulatory role of KLF15 in BCAA catabolism of muscle in Chinese soft-shelled turtles under nutritional deficiency, which might be activated the catabolism of BCAA in muscle to provide energy and maintain the homeostasis by KLF15-BACC signaling axis.
Collapse
Affiliation(s)
- Honghui Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Yaxiong Pan
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Lingsheng Bao
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Yulong Li
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Congyi Cheng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Li Liu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China; Hunan Fisheries Science Institute, Changsha 410153, China
| | - Jing Xiang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Jia Cheng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Jianshe Zhang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Wuying Chu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China.
| | - Yudong Shen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
41
|
Li H, An X, Bao L, Li Y, Pan Y, He J, Liu L, Zhu X, Zhang J, Cheng J, Chu W. MiR-125a-3p-KLF15-BCAA Regulates the Skeletal Muscle Branched-Chain Amino Acid Metabolism in Nile Tilapia ( Oreochromis niloticus) During Starvation. Front Genet 2020; 11:852. [PMID: 32849831 PMCID: PMC7431957 DOI: 10.3389/fgene.2020.00852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
The branched-chain amino acids (BCAAs) play a key role in the energy metabolism of the muscle tissue and the Krüppel-like factor 15 (KLF15) as a transcription factor, which is a key regulator of BCAA metabolism in the skeletal muscle. This study assessed the effect of starvation for 0, 3, 7, and 15 days on BCAA metabolism in the skeletal muscle of Nile tilapia. The results showed that the expression of KLF15 showed a trend of increasing first and then decreasing during starvation, as well as the expression and activity of branched-chain aminotransferase 2 (BCAT2) and alanine aminotransferase (ALT). On the other hand, the content of BCAA was at first decreased and then upregulated, and it reached the lowest level after starvation for 3 days. In addition, through dual-luciferase reporter assay and injection experiments, it was found that KLF15 is the target gene of miR-125a-3p, which further verified that miR-125a-3p can regulate the BCAA metabolism by targeting KLF15 in the skeletal muscle. Thus, our work investigated the possible mechanisms of BCAA metabolism adapting to nutritional deficiency in the skeletal muscle of Nile tilapia and illustrated the regulation of BCAA metabolism through the miR-125a-3p-KLF15-BCAA pathway in the skeletal muscle.
Collapse
Affiliation(s)
- Honghui Li
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Xiaoling An
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Lingsheng Bao
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Yulong Li
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Yaxiong Pan
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Jinggang He
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Li Liu
- Hunan Fisheries Science Institute, Changsha, China
| | - Xin Zhu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Jianshe Zhang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Jia Cheng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Wuying Chu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, College of Biological and Environmental Engineering, Changsha University, Changsha, China
| |
Collapse
|
42
|
Noack C, Iyer LM, Liaw NY, Schoger E, Khadjeh S, Wagner E, Woelfer M, Zafiriou MP, Milting H, Sossalla S, Streckfuss-Boemeke K, Hasenfuß G, Zimmermann WH, Zelarayán LC. KLF15-Wnt-Dependent Cardiac Reprogramming Up-Regulates SHISA3 in the Mammalian Heart. J Am Coll Cardiol 2020; 74:1804-1819. [PMID: 31582141 DOI: 10.1016/j.jacc.2019.07.076] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/31/2019] [Accepted: 07/12/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND The combination of cardiomyocyte (CM) and vascular cell (VC) fetal reprogramming upon stress culminates in end-stage heart failure (HF) by mechanisms that are not fully understood. Previous studies suggest KLF15 as a key regulator of CM hypertrophy. OBJECTIVES This study aimed to characterize the impact of KLF15-dependent cardiac transcriptional networks leading to HF progression, amenable to therapeutic intervention in the adult heart. METHODS Transcriptomic bioinformatics, phenotyping of Klf15 knockout mice, Wnt-signaling-modulated hearts, and pressure overload and myocardial ischemia models were applied. Human KLF15 knockout embryonic stem cells and engineered human myocardium, and human samples were used to validate the relevance of the identified mechanisms. RESULTS The authors identified a sequential, postnatal transcriptional repression mediated by KLF15 of pathways implicated in pathological tissue remodeling, including distinct Wnt-pathways that control CM fetal reprogramming and VC remodeling. The authors further uncovered a vascular program induced by a cellular crosstalk initiated by CM, characterized by a reduction of KLF15 and a concomitant activation of Wnt-dependent transcriptional signaling. Within this program, a so-far uncharacterized cardiac player, SHISA3, primarily expressed in VCs in fetal hearts and pathological remodeling was identified. Importantly, the KLF15 and Wnt codependent SHISA3 regulation was demonstrated to be conserved in mouse and human models. CONCLUSIONS The authors unraveled a network interplay defined by KLF15-Wnt dynamics controlling CM and VC homeostasis in the postnatal heart and demonstrated its potential as a cardiac-specific therapeutic target in HF. Within this network, they identified SHISA3 as a novel, evolutionarily conserved VC marker involved in pathological remodeling in HF.
Collapse
Affiliation(s)
- Claudia Noack
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany; Research & Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Lavanya M Iyer
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany; Computational and Systems Biology, Genome Institute of Singapore (GIS), Singapore
| | - Norman Y Liaw
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany
| | - Eric Schoger
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany
| | - Sara Khadjeh
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany; Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany
| | - Eva Wagner
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany; Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany
| | - Monique Woelfer
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany
| | - Maria-Patapia Zafiriou
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Samuel Sossalla
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany; Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Katrin Streckfuss-Boemeke
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany; Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany
| | - Gerd Hasenfuß
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany; Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany
| | - Laura C Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany.
| |
Collapse
|
43
|
Gottlieb RA, Bhowmick NA. Pushing the Heart Over a KLF(15). J Am Coll Cardiol 2020; 74:1820-1822. [PMID: 31582142 DOI: 10.1016/j.jacc.2019.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/14/2019] [Accepted: 08/20/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Roberta A Gottlieb
- Department of Medicine, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Neil A Bhowmick
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Department of Medicine, Cedars-Sinai Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
44
|
Li L, Li H, Tien CL, Jain MK, Zhang L. Kruppel-Like Factor 15 Regulates the Circadian Susceptibility to Ischemia Reperfusion Injury in the Heart. Circulation 2020; 141:1427-1429. [PMID: 32339045 PMCID: PMC7197441 DOI: 10.1161/circulationaha.119.041664] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Le Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77004, USA
- Department of Anesthesiology, Zhujiang hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Hui Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77004, USA
| | - Chih-Liang Tien
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77004, USA
| | - Mukesh K. Jain
- Case Cardiovascular Research Institute, Department of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
- School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77004, USA
| |
Collapse
|
45
|
Regulatory Mechanisms of Mitochondrial Function and Cardiac Aging. Int J Mol Sci 2020; 21:ijms21041359. [PMID: 32085438 PMCID: PMC7072955 DOI: 10.3390/ijms21041359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 01/10/2023] Open
Abstract
Aging is a major risk factor for cardiovascular diseases (CVDs), the major cause of death worldwide. Cardiac myocytes, which hold the most abundant mitochondrial population, are terminally differentiated cells with diminished regenerative capacity in the adult. Cardiomyocyte mitochondrial dysfunction is a characteristic feature of the aging heart and one out of the nine features of cellular aging. Aging and cardiac pathologies are also associated with increased senescence in the heart. However, the cause and consequences of cardiac senescence during aging or in cardiac pathologies are mostly unrecognized. Further, despite recent advancement in anti-senescence therapy, the targeted cell type and the effect on cardiac structure and function have been largely overlooked. The unique cellular composition of the heart, and especially the functional properties of cardiomyocytes, need to be considered when designing therapeutics to target cardiac aging. Here we review recent findings regarding key factors regulating cell senescence, mitochondrial health as well as cardiomyocyte rejuvenation.
Collapse
|
46
|
Liu B, Zhang TN, Knight JK, Goodwin JE. The Glucocorticoid Receptor in Cardiovascular Health and Disease. Cells 2019; 8:cells8101227. [PMID: 31601045 PMCID: PMC6829609 DOI: 10.3390/cells8101227] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022] Open
Abstract
The glucocorticoid receptor is a member of the nuclear receptor family that controls many distinct gene networks, governing various aspects of development, metabolism, inflammation, and the stress response, as well as other key biological processes in the cardiovascular system. Recently, research in both animal models and humans has begun to unravel the profound complexity of glucocorticoid signaling and convincingly demonstrates that the glucocorticoid receptor has direct effects on the heart and vessels in vivo and in vitro. This research has contributed directly to improving therapeutic strategies in human disease. The glucocorticoid receptor is activated either by the endogenous steroid hormone cortisol or by exogenous glucocorticoids and acts within the cardiovascular system via both genomic and non-genomic pathways. Polymorphisms of the glucocorticoid receptor are also reported to influence the progress and prognosis of cardiovascular disease. In this review, we provide an update on glucocorticoid signaling and highlight the critical role of this signaling in both physiological and pathological conditions of the cardiovascular system. With increasing in-depth understanding of glucocorticoid signaling, the future is promising for the development of targeted glucocorticoid treatments and improved clinical outcomes.
Collapse
Affiliation(s)
- Bing Liu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Tie-Ning Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jessica K Knight
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
47
|
Svenson KL, Long LL, Ciciotte SL, Adams MD. A mutation in mouse Krüppel-like factor 15 alters the gut microbiome and response to obesogenic diet. PLoS One 2019; 14:e0222536. [PMID: 31553739 PMCID: PMC6760833 DOI: 10.1371/journal.pone.0222536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/30/2019] [Indexed: 12/22/2022] Open
Abstract
We identified a mouse strain, HLB444, carrying an N-ethyl-N-nitrosourea (ENU)-induced mutation in a highly conserved C2H2 zinc-finger DNA binding motif of the transcriptional regulator KLF15 that exhibits resistance to diet-induced obesity. Characterization of the HLB444 mutant model on high-fat and chow diets revealed a number of phenotypic differences compared to wild-type controls. When fed a high fat diet, HLB444 had lower body fat, resistance to hepatosteatosis, lower circulating glucose and improved insulin sensitivity compared to C57BL/6J controls. Gut microbial profiles in HLB444 generated from 16S rRNA sequencing of fecal samples differed from controls under both chow and high fat diets. HLB444 shares similar phenotypic traits with engineered full- and adipose-specific Klf15 knockout strains; however, some phenotypic differences between this mutant and the other models suggest that the Klf15 mutation in HLB444 is a hypomorphic variant. The HLB444 model will inform further annotation of transcriptional functions of KLF15, especially with respect to the role of the first zinc-finger domain.
Collapse
Affiliation(s)
- Karen L. Svenson
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Lauren L. Long
- The Jackson Laboratory, Farmington, Connecticut, United States of America
| | | | - Mark D. Adams
- The Jackson Laboratory, Farmington, Connecticut, United States of America
| |
Collapse
|
48
|
Zhao Z, Tian H, Shi B, Jiang Y, Liu X, Hu J. Transcriptional Regulation of the Bovine Fatty Acid Transport Protein 1 Gene by Krüppel-Like Factors 15. Animals (Basel) 2019; 9:ani9090654. [PMID: 31491871 PMCID: PMC6769441 DOI: 10.3390/ani9090654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 12/27/2022] Open
Abstract
Simple Summary The nutritional value and qualities of beef are enhanced when the unsaturated fatty acid content is increased. Fatty acid transport protein 1 (FATP1), also called SLC27A1, an integral membrane protein that facilitates long-chain fatty acid influx, is involved in the genetic network for oleic acid synthesis in beef. Polymorphisms in bovine SLC27A1 gene are most significantly associated with oleic acid. Its expression exhibits significant positive correlations with bovine intramuscular fat content in the longissimus thoracis muscle. However, the transcription factors that contribute to the control and regulation of its expression have not been characterized extensively. In this study, we determined the tissue distribution of SLC27A1 mRNA and found that bovine SLC27A1 was highly expressed in subcutaneous adipose tissue and the longissimus thoracis muscle. Furthermore, we analyzed the molecular mechanisms involved in SLC27A1 regulation and found that the transcriptional activity of SLC27A1 gene was dependent on KLF15 transcription factor. These results may lead to an enhanced understanding of the regulation of SLC27A1 expression in other models, as well as provide new insights into the regulatory mechanism and biological functions of the SLC27A1 gene in determining the lipid composition in beef. Abstract Oleic acid is a major monounsaturated fatty acid, which accounts for about 33% of the fatty acid content in beef and is considered to have the least negative effect on serum cholesterol levels. Fatty acid transport protein 1 (FATP1), an integral membrane protein that facilitates long-chain fatty acid (LCFA) influx, is involved in the genetic network for oleic acid synthesis in beef. Its expression exhibits significant positive correlations with intramuscular fat (IMF) content in the longissimus thoracis. However, the expression mechanism of SLC27A1 or FATP1 is still unclear. To elucidate the molecular mechanisms involved in bovine SLC27A1 regulation, we cloned and characterized the promoter region of SLC27A1. By applying 5′-rapid amplification of cDNA end analysis, we identified two alternative splice variants of this gene. Using a series of 5′ deletion promoter plasmids in luciferase reporter assays, we found that the core promoter was 96 base pairs upstream from the transcription initiation site. Electrophoretic mobility shift assay combined with a site-directed mutation experiment demonstrated that KLF15 binding to the promoter region drives the SLC27A1 transcription. KLF15 plays an essential role in adipogenesis and skeletal muscle lipid flux. Thus, these results might provide further information on the regulatory roles of SLC27A1 gene in mediating the lipid composition in beef.
Collapse
Affiliation(s)
- Zhidong Zhao
- College of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Hongshan Tian
- College of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Bingang Shi
- College of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Yanyan Jiang
- College of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Xiu Liu
- College of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Jiang Hu
- College of Animal Science and Technology & Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| |
Collapse
|
49
|
Hafidi ME, Buelna-Chontal M, Sánchez-Muñoz F, Carbó R. Adipogenesis: A Necessary but Harmful Strategy. Int J Mol Sci 2019; 20:ijms20153657. [PMID: 31357412 PMCID: PMC6696444 DOI: 10.3390/ijms20153657] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/16/2019] [Accepted: 07/20/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity is considered to significantly increase the risk of the development of a vast range of metabolic diseases. However, adipogenesis is a complex physiological process, necessary to sequester lipids effectively to avoid lipotoxicity in other tissues, like the liver, heart, muscle, essential for maintaining metabolic homeostasis and has a crucial role as a component of the innate immune system, far beyond than only being an inert mass of energy storage. In pathophysiological conditions, adipogenesis promotes a pro-inflammatory state, angiogenesis and the release of adipokines, which become dangerous to health. It results in a hypoxic state, causing oxidative stress and the synthesis and release of harmful free fatty acids. In this review, we try to explain the mechanisms occurring at the breaking point, at which adipogenesis leads to an uncontrolled lipotoxicity. This review highlights the types of adipose tissue and their functions, their way of storing lipids until a critical point, which is associated with hypoxia, inflammation, insulin resistance as well as lipodystrophy and adipogenesis modulation by Krüppel-like factors and miRNAs.
Collapse
Affiliation(s)
- Mohammed El Hafidi
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico
| | - Mabel Buelna-Chontal
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología "Ignacio Chávez", México City 14080, Mexico.
| |
Collapse
|
50
|
Uddin GM, Zhang L, Shah S, Fukushima A, Wagg CS, Gopal K, Al Batran R, Pherwani S, Ho KL, Boisvenue J, Karwi QG, Altamimi T, Wishart DS, Dyck JRB, Ussher JR, Oudit GY, Lopaschuk GD. Impaired branched chain amino acid oxidation contributes to cardiac insulin resistance in heart failure. Cardiovasc Diabetol 2019; 18:86. [PMID: 31277657 PMCID: PMC6610921 DOI: 10.1186/s12933-019-0892-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/28/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Branched chain amino acids (BCAA) can impair insulin signaling, and cardiac insulin resistance can occur in the failing heart. We, therefore, determined if cardiac BCAA accumulation occurs in patients with dilated cardiomyopathy (DCM), due to an impaired catabolism of BCAA, and if stimulating cardiac BCAA oxidation can improve cardiac function in mice with heart failure. METHOD For human cohorts of DCM and control, both male and female patients of ages between 22 and 66 years were recruited with informed consent from University of Alberta hospital. Left ventricular biopsies were obtained at the time of transplantation. Control biopsies were obtained from non-transplanted donor hearts without heart disease history. To determine if stimulating BCAA catabolism could lessen the severity of heart failure, C57BL/6J mice subjected to a transverse aortic constriction (TAC) were treated between 1 to 4-week post-surgery with either vehicle or a stimulator of BCAA oxidation (BT2, 40 mg/kg/day). RESULT Echocardiographic data showed a reduction in ejection fraction (54.3 ± 2.3 to 22.3 ± 2.2%) and an enhanced formation of cardiac fibrosis in DCM patients when compared to the control patients. Cardiac BCAA levels were dramatically elevated in left ventricular samples of patients with DCM. Hearts from DCM patients showed a blunted insulin signalling pathway, as indicated by an increase in P-IRS1ser636/639 and its upstream modulator P-p70S6K, but a decrease in its downstream modulators P-AKT ser473 and in P-GSK3β ser9. Cardiac BCAA oxidation in isolated working hearts was significantly enhanced by BT2, compared to vehicle, following either acute or chronic treatment. Treatment of TAC mice with BT2 significantly improved cardiac function in both sham and TAC mice (63.0 ± 1.8 and 56.9 ± 3.8% ejection fraction respectively). Furthermore, P-BCKDH and BCKDK expression was significantly decreased in the BT2 treated groups. CONCLUSION We conclude that impaired cardiac BCAA catabolism and insulin signaling occur in human heart failure, while enhancing BCAA oxidation can improve cardiac function in the failing mouse heart.
Collapse
Affiliation(s)
- Golam M Uddin
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - Liyan Zhang
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - Saumya Shah
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - Arata Fukushima
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - Cory S Wagg
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - Keshav Gopal
- Katz Centre for Pharmacy and Health Research, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Rami Al Batran
- Katz Centre for Pharmacy and Health Research, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Simran Pherwani
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - Kim L Ho
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada.,Katz Centre for Pharmacy and Health Research, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Jamie Boisvenue
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada.,Department of Pharmacology, College of Medicine, University of Diyala, Diyala, Iraq
| | - Tariq Altamimi
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada
| | - David S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Canada.,Metabolomics Innovation Centre, University of Alberta, Edmonton, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - John R Ussher
- Katz Centre for Pharmacy and Health Research, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.,Divsion of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, 423 Heritage Medical Research Centre, Edmonton, T6G 2S2, Canada. .,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada. .,Alberta Diabetes Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|