1
|
Zaccagnini G, Baci D, Tastsoglou S, Cozza I, Madè A, Voellenkle C, Nicoletti M, Ruatti C, Longo M, Perani L, Gaetano C, Esposito A, Martelli F. miR-210 overexpression increases pressure overload-induced cardiac fibrosis. Noncoding RNA Res 2025; 12:20-33. [PMID: 40034123 PMCID: PMC11874870 DOI: 10.1016/j.ncrna.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
Aortic stenosis, a common valvular heart disease, can lead to left ventricular pressure overload, triggering pro-fibrotic responses in the heart. miR-210 is a microRNA that responds to hypoxia and ischemia and plays a role in immune regulation and in cardiac remodeling upon myocardial infarction. This study investigated the effects of miR-210 on cardiac fibrosis caused by pressure overload. Using a mouse model with inducible miR-210 over-expression, we subjected mice to transverse aortic constriction (TAC) to induce pressure overload. Mice with miR-210 over-expression developed eccentric hypertrophy, heightened expression of hypertrophic markers (Nppa and Nppb) and increased cross sectional area of cardiomyocytes, impacting the free wall of the left ventricle. These findings suggest that miR-210 worsens cardiac dysfunction. Furthermore, miR-210 over-expression led to a more robust and sustained inflammatory response in the heart, increased interstitial and perivascular fibrosis, and activation of myofibroblasts. miR-210 also promoted angiogenesis. In vitro, cardiac fibroblasts over-expressing miR-210 showed increased adhesion, wound healing and migration capacity. Our results demonstrate that miR-210 contributes to adverse cardiac remodeling in response to pressure overload, including eccentric hypertrophy, inflammation, and fibrosis.
Collapse
Affiliation(s)
- G. Zaccagnini
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - D. Baci
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - S. Tastsoglou
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - I. Cozza
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - A. Madè
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - C. Voellenkle
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - M. Nicoletti
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - C. Ruatti
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - M. Longo
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - L. Perani
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - C. Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | - A. Esposito
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - F. Martelli
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
2
|
He L, She X, Guo L, Gao M, Wang S, Lu Z, Guo H, Li R, Nie Y, Xing J, Ji L. Hepatic AKAP1 deficiency exacerbates diet-induced MASLD by enhancing GPAT1-mediated lysophosphatidic acid synthesis. Nat Commun 2025; 16:4286. [PMID: 40341440 PMCID: PMC12062205 DOI: 10.1038/s41467-025-58790-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/02/2025] [Indexed: 05/10/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), closely associated with obesity, can progress to metabolic dysfunction-associated steatohepatitis when the liver undergoes overt inflammatory damage. A-kinase anchoring protein 1 (AKAP1) has been shown to control lipid accumulation in brown adipocytes. However, the role of AKAP1 signaling in hepatic lipid metabolism and MASLD remains poorly understood. Here, we showed that hepatocyte-specific AKAP1 deficiency exacerbated hepatic steatosis and steatohepatitis in male mice subjected to a high-fat diet and fast-food diet, respectively. Mechanistically, AKAP1 directly phosphorylated and inactivated glycerol-3-phosphate acyltransferase 1 (GPAT1) in a PKA-dependent manner, thus suppressing lysophosphatidic acid (LPA) production. Increased endogenous LPA in hepatocytes promoted hepatocellular triglyceride (TG) synthesis and initiated pronounced inflammatory response in Kupffer cells. Restoring hepatic AKAP1 or repressing LPA levels via GPAT1 knockdown alleviated MASLD exacerbation. Overall, AKAP1 plays a protective role against MASLD by inhibiting GPAT1 activity, highlighting the potential of targeting AKAP1/PKA/GPAT1 signalosome for MASLD therapy.
Collapse
Affiliation(s)
- Linjie He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaojuan She
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
- College of Life Sciences, Yan'an University, Yan'an, Shaanxi, China
| | - Lifei Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
- College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Mingshu Gao
- College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- National Demonstration Center for Experimental Basic Medical Science Education, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuangbin Wang
- National Demonstration Center for Experimental Basic Medical Science Education, Fourth Military Medical University, Xi'an, Shaanxi, China
- Medical College of Yan'an University, Yan'an, Shaanxi, China
| | - Zhenxing Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haitao Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Renlong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinliang Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Lele Ji
- National Demonstration Center for Experimental Basic Medical Science Education, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Wu Z, Huang N, Li C, Lin M, Chen Z, Li W, Zhou H. β-Sitosterol suppresses NLRP3 Inflammasome activation and Pyroptosis in myocardial ischemia/reperfusion injury via inhibition of PPARγ2. Int Immunopharmacol 2025; 154:114543. [PMID: 40157084 DOI: 10.1016/j.intimp.2025.114543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND β-Sitosterol, a plant-derived sterol, has demonstrated potential therapeutic effects in cardiovascular diseases, particularly myocardial ischemia-reperfusion injury (MIRI). Our study investigates its underlying mechanism through regulation of pyroptosis. METHODS To understand the role of β-sitosterol in protecting cardiomyocytes, MIRI rats were treated with β-sitosterol. Rats' cardiac functions were monitored, and hearts were harvested for histology and Western Blot analysis. Immunofluorescence, immunoblot, enzyme-linked immunosorbent assay, as well as overexpression and knockdown techniques were utilized in this study to investigate the molecular mechanisms underlying the cardioprotective effects of β-sitosterol. RESULTS Our results showed that β-Sitosterol significantly reduced H/R-induced pyroptosis in cardiomyocytes by decreasing cleaved caspase-1, gasdermin D (GSDMD), interleukin-1β (IL-1β), and interleukin-18 (IL-18). Immunofluorescence staining confirmed suppression of NLRP3 inflammasome activation. Notably, β-Sitosterol inhibited pyroptosis induced by ATP and ATP/LPS through the regulation of PPARγ2. Moreover, PPARγ2 upregulation promoted ATP and ATP/LPS-induced pyroptosis through the NLRP3/caspase-1/GSDMD pathway. In vivo, β-sitosterol alleviates myocardial ischemia-reperfusion injury-induced cardiac dysfunction and myocardial fibrosis in rats. CONCLUSIONS These findings provide new evidence supporting β-sitosterol as a potential therapeutic agent for cardiovascular diseases involving ischemic injury. Its protective effects may be mediated through targeting PPARγ2 and modulating NLRP3-dependent pyroptosis.
Collapse
Affiliation(s)
- Zheyi Wu
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China; Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Niwen Huang
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chao Li
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Muzhi Lin
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhangrong Chen
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Li
- Clinical Medical College, Guizhou Medical University, Guiyang, China.
| | - Haiyan Zhou
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
4
|
Liu T, Li P, Ou Z, Feng Y, Wang B, Yu T, Zhu Y, Yu L. Insights into the special physiology of Mortierella alpina cultured by agar supported solid state fermentation in enhancing arachidonic acid enriched lipid production. Sci Rep 2025; 15:15967. [PMID: 40335580 PMCID: PMC12058991 DOI: 10.1038/s41598-025-00965-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Solid-state fermentation (SSF), an eco-friendly technology, has shown the high-yield ability to produce products such as biodiesel, pharmaceuticals, and enzymes. However, it has not yet demonstrated an advantage in ARA-containing lipids production. This study demonstrated that agar-supported SSF (AgSF) could induce Mortierella alpina M0223 to yield higher ARA-rich lipids than submerged fermentation (SmF), and elucidated the underlying mechanisms by the comparative transcriptome. AgSF-M0223 formed a mycelial network consisting mainly of surface (SH) and aerial hyphae (AH). The attenuated citrate cycle of SH compared to SmF was coupled with enhanced triglyceride biosynthesis, glycerophospholipid metabolism, and underlying increases in NADPH supply, prompting more glucose flux towards ARA-rich lipid synthesis. Besides, AH has high initial lipid and ARA amounts, while its primary metabolism was weakened due to nutrient scarcity, demonstrating attenuated lipid synthesis. The unique ARA and lipid synthesis characteristics of SH and AH enabled AgSF-M0223 to achieve high-yield ARA-rich lipids. By supplementing nutrients to AH through a spraying strategy and optimizing nutrients for SH, lipid yields reached 12.64 g/L comprising 70.41% ARA, 1.63 times higher than before optimization. These findings provided new insights into fungal physiology under SSF, and presented a promising eco-friendly paradigm for ARA production with advances in mechanical automation.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Food and Medicine Resources Engineering Research Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Pandeng Li
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ziqi Ou
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yumei Feng
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Food and Medicine Resources Engineering Research Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bohan Wang
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Food and Medicine Resources Engineering Research Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Tianyi Yu
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Molecular Biophysics, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yuanmin Zhu
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Key Laboratory of Molecular Biophysics, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Hubei Food and Medicine Resources Engineering Research Center, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Longjiang Yu
- Department of Biotechnology, College of Life Science and Technology, Institute of Resource Biology and Biotechnology, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Key Laboratory of Molecular Biophysics, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Hubei Food and Medicine Resources Engineering Research Center, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
5
|
Hou YJ, Yang XX, Meng HX. Mitochondrial metabolism in laryngeal cancer: therapeutic mechanisms and prospects. Biochim Biophys Acta Rev Cancer 2025; 1880:189335. [PMID: 40311711 DOI: 10.1016/j.bbcan.2025.189335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
Tumours reprogram pathways that regulate nutrient uptake and metabolism to meet the biosynthetic, bioenergetic, and redox requirements of cancer cells. This phenomenon is known as metabolic reprogramming and is edited by the deletion of oncogenes and the activation of proto-oncogenes. This article highlights the pathological mechanisms associated with metabolic reprogramming in laryngeal cancer (LC), including enhanced glycolysis, tricarboxylic acid cycle, nucleotide synthesis, lipid synthesis and metabolism, and amino acid metabolism, with a special emphasis on glutamine, tryptophan, and arginine metabolism. All these changes are regulated by HPV infection, hypoxia, and metabolic mediators in the tumour microenvironment. We analyzed the function of metabolic reprogramming in the development of drug resistance during standard LC treatment, which is challenging. In addition, we revealed recent advances in targeting metabolic strategies, assessing the strengths and weaknesses of clinical trials and treatment programs to attack resistance. This review summarises some currently important biomarkers and lays the foundation for therapeutic pathways in LC.
Collapse
Affiliation(s)
- Yun-Jing Hou
- Harbin Medical University, Harbin, China; Harbin Medical University Cancer Hospital, Harbin, China; Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin-Xin Yang
- Harbin Medical University, Harbin, China; Harbin Medical University Cancer Hospital, Harbin, China; Department of Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Xue Meng
- Harbin Medical University, Harbin, China; Harbin Medical University Cancer Hospital, Harbin, China; Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
6
|
Oyamada H, Ying Y, Agrawal S, Liu A, Subramanian VS, de Melo Bento CA, Agrawal A. Chronic IL-21 drives neuroinflammation and promotes lipid accumulation in microglia. Immun Ageing 2025; 22:15. [PMID: 40301833 PMCID: PMC12039274 DOI: 10.1186/s12979-025-00510-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
Neuroinflammation is a key contributor to the onset and progression of neurodegenerative diseases, driven by factors such as viral infections, autoimmune disorders, and peripheral inflammation. However, the mechanisms linking peripheral inflammation or viral infections to neuroinflammation remain poorly understood, limiting the development of effective therapies. Proinflammatory cytokines are implicated in these processes but their effects on brain cells, including microglia, remain insufficiently characterized. Here, we demonstrate that IL-21, a proinflammatory cytokine elevated in autoimmune disorders, chronic viral infections, and Alzheimer's disease, activates microglia and promotes lipid accumulation within these cells. Young, healthy mice injected with IL-21 to mimic chronic exposure exhibited increased proinflammatory cytokine levels and microglial activation in the brain. Notably, microglia in these mice displayed enhanced lipid accumulation, accompanied by upregulation of lipid uptake receptors such as CD36 and TREM-2. These findings were corroborated using the human microglial cell line HMC-3, where IL-21 exposure similarly induced lipid accumulation and increased expression of CD36 and ApoE. Mechanistic investigations revealed that IL-21 upregulates HIF-1α, a transcription factor critical for lipid metabolism and lipid droplet formation. Additionally, we observed elevated IL-21 levels in the circulation of elderly individuals compared to younger counterparts, with IL-21 increases associated with CMV seropositivity. Aged mouse brains mirrored the microglial lipid accumulation and activation patterns seen in IL-21-injected mice. In summary, we identify a novel IL-21-driven mechanism involving lipid accumulation in microglia that contributes to neuroinflammation.
Collapse
Affiliation(s)
- Hugo Oyamada
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Yinzhi Ying
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Aizhu Liu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Veedamali S Subramanian
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Cleonice Alves de Melo Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
7
|
Li H, Xiao F, Zhou C, Zhu T, Wang S. Metabolic Adaptations and Therapies in Cardiac Hypoxia: Mechanisms and Clinical Implications/ Potential Strategies. JACC Basic Transl Sci 2025:S2452-302X(24)00458-3. [PMID: 40265246 DOI: 10.1016/j.jacbts.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 04/24/2025]
Abstract
Cardiac hypoxia triggers a cascade of responses and functional changes in myocardial and non-myocardial cells, profoundly affecting cellular metabolism, oxygen-sensing mechanisms, and immune responses. Myocardial cells, being the primary cell type in cardiac tissue, undergo significant alterations in energy metabolism, including glycolysis, fatty acid metabolism, ketone body utilization, and branched-chain amino acid metabolism, to maintain cardiac function under hypoxic conditions. Non-myocardial cells, such as fibroblasts, endothelial cells, and immune cells, although fewer in number, play crucial roles in regulating cardiac homeostasis, maintaining structural integrity, and responding to injury. This review discusses the metabolic reprogramming of immune cells, particularly macrophages, during ischemia-reperfusion injury and explores various therapeutic strategies that modulate these metabolic pathways to protect the heart during hypoxia. Understanding these interactions provides valuable insights and potential therapeutic targets for heart disease treatment.
Collapse
Affiliation(s)
- Huili Li
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fei Xiao
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenghui Zhou
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012, West China Hospital, Sichuan University, Chengdu, China.
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Linzhi People's Hospital, Linzhi, Tibet, China.
| |
Collapse
|
8
|
Wang M, Lu R, Peng L, Xu LL, He SF, Guo T, Lu MJ, Luo Y, Cui TT. MICRORNA-146B TARGETS HIF-1Α AND ATTENUATES CARDIOMYOCYTE APOPTOSIS AND FIBROSIS IN DOXORUBICIN-INDUCED HEART FAILURE. Shock 2025; 63:656-663. [PMID: 39874498 PMCID: PMC11939110 DOI: 10.1097/shk.0000000000002546] [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: 06/22/2024] [Revised: 07/18/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025]
Abstract
ABSTRACT The global prevalence of heart failure is still growing, which imposes a heavy economic burden. The role of microRNA-146b (miR-146b) in HF remains largely unknown. This study aims to explore the role and mechanism of miR-146b in HF. Method: We applied reverse transcription-polymerase chain reaction to search for differential microRNAs between myocardial tissues of heart failure patients and controls. We also used reverse transcription-polymerase chain reaction to detect the miR-146b expression in primary neonatal mouse cardiomyocytes and mice models of doxorubicin-induced HF. In vivo experiments, echocardiography was performed at baseline and weeks 6. After that we harvested mice's heart and evaluated the cardiomyocyte with hematoxylin and eosin (HE), Masson trichrome staining, and TUNEL staining. Through bioinformatics analysis, we found HIF-1α might be the target gene of miR-146b, which validated by luciferase reporter gene assay. Subsequently, mRNA and protein expression levels of HIF-1α were detected by overexpression or inhibition of miR-146b in primary neonatal mouse cardiomyocytes. Results: We found that miR-146b expression was decreased in myocardial tissues of HF patients compared with controls ( P < 0.01). MiR-146b levels were notably downregulated in HF models. MiR-146b knockout mice showed a more pronounced decrease in cardiac function and more severe myocardial fibrosis and apoptosis than wild type. Meanwhile, over expression or repression of miR-146b in primary neonatal mouse cardiomyocytes could inhibit or upregulate HIF-1α mRNA and protein expression. Conclusion : Our study shows that miR-146b may be a protective factor for cardiomyocytes by modulating HIF-1α.
Collapse
Affiliation(s)
- Min Wang
- Jinan University, Guangzhou, China
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rui Lu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liang Peng
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling-Ling Xu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shang-Fei He
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tao Guo
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ming-Jun Lu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yi Luo
- Jinan University, Guangzhou, China
- Department of Cardiology, Guangzhou First People’s Hospital, Guangzhou, China
| | - Tong-Tao Cui
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Jiang M, Huang J, Guo X, Fu W, Peng L, Wang Y, Liu W, Liu J, Zhou L, Xiao Y. HIF-3α/PPAR-γ Regulates Hypoxia Tolerance by Altering Glycolysis and Lipid Synthesis in Blunt Snout Bream ( Megalobrama amblycephala). Int J Mol Sci 2025; 26:2613. [PMID: 40141255 PMCID: PMC11942064 DOI: 10.3390/ijms26062613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Hypoxic stress causes cell damage and serious diseases in organisms, especially in aquatic animals. It is important to elucidate the changes in metabolic function caused by hypoxia and the mechanisms underlying these changes. This study focuses on the low oxygen tolerance feature of a new blunt snout bream strain (GBSBF1). Our data show that GBSBF1 has a different lipid and carbohydrate metabolism pattern than wild-type bream, with altering glycolysis and lipid synthesis. In GBSBF1, the expression levels of phd2 and vhl genes are significantly decreased, while the activation of HIF-3α protein is observed to have risen significantly. The results indicate that enhanced HIF-3α can positively regulate gpd1ab and gpam through PPAR-γ, which increases glucose metabolism and reduces lipolysis of GBSBF1. This research is beneficial for creating new aquaculture strains with low oxygen tolerance traits.
Collapse
Affiliation(s)
- Minggui Jiang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jing Huang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Xing Guo
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Wen Fu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Liangyue Peng
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Wenbin Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jinhui Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Yamei Xiao
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| |
Collapse
|
10
|
Lindsay RT, Rhodes CJ. Reactive Oxygen Species (ROS) in Metabolic Disease-Don't Shoot the Metabolic Messenger. Int J Mol Sci 2025; 26:2622. [PMID: 40141264 PMCID: PMC11942130 DOI: 10.3390/ijms26062622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Reactive oxygen species (ROS) are widely considered key to pathogenesis in chronic metabolic disease. Consequently, much attention is rightly focused on minimising oxidative damage. However, for ROS production to be most effectively modulated, it is crucial to first appreciate that ROS do not solely function as pathological mediators. There are >90 gene products specifically evolved to generate, handle, and tightly buffer the cellular concentration of ROS. Therefore, it is likely that ROS plays a role as integral homeostatic signalling components and only become toxic in extremis. This review explores these commonly overlooked normal physiological functions, including how ROS are generated in response to environmental or hormonal stimuli, the mechanisms by which the signals are propagated and regulated, and how the cell effectively brings the signal to an end after an appropriate duration. In the course of this, several specific and better-characterised signalling mechanisms that rely upon ROS are explored, and the threshold at which ROS cross from beneficial signalling molecules to pathology mediators is discussed.
Collapse
Affiliation(s)
- Ross T. Lindsay
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Christopher J. Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| |
Collapse
|
11
|
Wang Y, Wang Y, Zhang W. Dysregulation of Mitochondrial in Pulmonary Hypertension-Related Right Ventricular Remodeling: Pathophysiological Features and Targeting Drugs. Rev Cardiovasc Med 2025; 26:25781. [PMID: 40160582 PMCID: PMC11951289 DOI: 10.31083/rcm25781] [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: 07/21/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 04/02/2025] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening condition characterized by right ventricular (RV) remodeling, which is a major determinant of patient survival. The progression of right ventricular remodeling is significantly influenced by mitochondrial dysfunction, providing profound insights into vascular health and cardiovascular risk. In this review, we discuss the molecular targets, pathophysiological characteristics, and potential mechanisms underlying mitochondrial dysfunction in PH, encompassing disturbances in mitochondrial dynamics, inflammation, and dysregulation of mitochondrial energy metabolism. Finally, we review the primary therapeutic targets currently utilized to address cardiac dysfunction resulting from mitochondrial damage. Hopefully, this might inspire novel approaches to the management of cardiovascular disorders.
Collapse
Affiliation(s)
- Yuehan Wang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Yingzhuo Wang
- The First Clinical Medical College, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Weifang Zhang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| |
Collapse
|
12
|
Reddan B, Cummins EP. The regulation of cell metabolism by hypoxia and hypercapnia. J Biol Chem 2025; 301:108252. [PMID: 39914740 PMCID: PMC11923829 DOI: 10.1016/j.jbc.2025.108252] [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: 09/18/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 03/06/2025] Open
Abstract
Every cell in the body is exposed to a certain level of CO2 and O2. Hypercapnia and hypoxia elicit stress signals to influence cellular metabolism and function. Both conditions exert profound yet distinct effects on metabolic pathways and mitochondrial dynamics, highlighting the need for cells to adapt to changes in the gaseous microenvironment. The interplay between hypercapnia and hypoxia signaling is the key for dictating cellular homeostasis as microenvironmental CO2 and O2 levels are inextricably linked. Hypercapnia, characterized by elevated pCO2, introduces metabolic adaptations within the aerobic metabolism pathways, affecting tricarboxylic acid cycle flux, lipid, and amino acid metabolism, oxidative phosphorylation and the electron transport chain. Hypoxia, defined by reduced oxygen availability, necessitates a shift from oxidative phosphorylation to anaerobic glycolysis to sustain ATP production, a process orchestrated by the stabilization of hypoxia-inducible factor-1α. Given that hypoxia and hypercapnia are present in both physiological and cancerous microenvironments, how might the coexistence of hypercapnia and hypoxia influence metabolic pathways and cellular function in physiological niches and the tumor microenvironment?
Collapse
Affiliation(s)
- Ben Reddan
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Eoin P Cummins
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
13
|
Kim H, Park C, Wei X, Chhetri A, Manandhar L, Jang G, Hwang J, Chinbold B, Chuluunbaatar C, Kwon HM, Park R. Golgi condensation causes intestinal lipid accumulation through HIF-1α-mediated GM130 ubiquitination by NEDD4. Exp Mol Med 2025; 57:349-363. [PMID: 39900792 PMCID: PMC11873256 DOI: 10.1038/s12276-025-01396-2] [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: 03/12/2024] [Revised: 08/26/2024] [Accepted: 11/14/2024] [Indexed: 02/05/2025] Open
Abstract
The breakdown of Golgi proteins disrupts lipid trafficking, leading to lipid accumulation in the small intestine. However, the causal mechanism of the effects of Golgi protein degradation on the Golgi structure related to lipid trafficking in the small intestine remains unknown. Here we find that Golgi protein degradation occurs under hypoxic conditions in high-fat-diet-fed mice. Hypoxia-induced degradation promotes structural changes in the Golgi apparatus, termed 'Golgi condensation'. In addition, hypoxia-inducible factor 1α (HIF-1α) activation enhances Golgi condensation through the ubiquitination and degradation of Golgi matrix protein 130 (GM130), which is facilitated by neural precursor cell expressed developmentally downregulated protein 4 (NEDD4). Golgi condensation upon exposure to hypoxia promotes lipid accumulation, apolipoprotein A1 retention and decreased chylomicron secretion in the intestinal epithelium. Golgi condensation and lipid accumulation induced by GM130 depletion are reversed by exogenous GM130 induction in the intestinal epithelium. Inhibition of either HIF-1α or NEDD4 protects against GM130 degradation and, thereby, rescues cells from Golgi condensation, which further increases apolipoprotein A1 secretion and lipid accumulation both in vivo and in vitro. Furthermore, the HIF-1α inhibitor PX-478 prevents Golgi condensation, which decreases lipid accumulation and promotes high-density lipoprotein secretion in high-fat-diet-fed mice. Overall, our results suggest that Golgi condensation plays a key role in lipid trafficking in the small intestine through the HIF-1α- and NEDD4-mediated degradation of GM130, and these findings highlight the possibility that the prevention of structural modifications in the Golgi apparatus can ameliorate intestinal lipid accumulation in obese individuals.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Channy Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Xiaofan Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Arun Chhetri
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Laxman Manandhar
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Gyuho Jang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jaetaek Hwang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Batchingis Chinbold
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Chagtsalmaa Chuluunbaatar
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Hyug Moo Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| |
Collapse
|
14
|
Zhou F, Zhou JB, Wei TP, Wu D, Wang RX. The Role of HIF-1α in Atrial Fibrillation: Recent Advances and Therapeutic Potentials. Rev Cardiovasc Med 2025; 26:26787. [PMID: 40026494 PMCID: PMC11868874 DOI: 10.31083/rcm26787] [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: 09/29/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 03/05/2025] Open
Abstract
The steady increase in life expectancy throughout the world is contributing to an increased incidence of atrial fibrillation (AF), which imposes a significant socioeconomic toll on affected patients and societies. The mechanisms underlying atrial fibrillation are multifaceted and vary among individuals. Hypoxia is a process that is closely linked to AF onset and progression. Hypoxia-inducible factor 1-alpha (HIF-1α) is a transcription factor that serves as a key regulator of oxygen homeostasis within cells through its activation under hypoxic conditions and subsequently coordinates various pathophysiological responses. High levels of HIF-1α expression are evident in AF patients, and facilitate the progression from persistent AF to permanent AF. Thus, HIF-1α may serve as a promising target for novel therapeutic strategies aimed at the prevention and treatment of AF. This review provides an overview and synthesis of recent studies probing the relationship between HIF-1α and AF, providing a foundation for future studies and the development targeted drug therapies.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Jia-Bin Zhou
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Tian-Peng Wei
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Dan Wu
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| |
Collapse
|
15
|
Gonzalez GA, Osuji EU, Fiur NC, Clark MG, Ma S, Lukov LL, Zhang C. Alteration of Lipid Metabolism in Hypoxic Cancer Cells. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:25-34. [PMID: 39886224 PMCID: PMC11775851 DOI: 10.1021/cbmi.4c00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 02/01/2025]
Abstract
Due to uncontrolled cell proliferation and disrupted vascularization, many cancer cells in solid tumors have limited oxygen supply. The hypoxic microenvironments of tumors lead to metabolic reprogramming of cancer cells, contributing to therapy resistance and metastasis. To identify better targets for the effective removal of hypoxia-adaptive cancer cells, it is crucial to understand how cancer cells alter their metabolism in hypoxic conditions. Here, we studied lipid metabolic changes in cancer cells under hypoxia using coherent Raman scattering (CRS) microscopy. We discovered the accumulation of lipid droplets (LDs) in the endoplasmic reticulum (ER) in hypoxia. Time-lapse CRS microscopy revealed the release of old LDs and the reaccumulated LDs in the ER during hypoxia exposure. Additionally, we explored the impact of carbon sources on LD formation and found that MIA PaCa2 cells preferred fatty acid uptake for LD formation, while glucose was essential to alleviate lipotoxicity. Hyperspectral-stimulated Raman scattering (SRS) microscopy revealed a reduction in cholesteryl ester content and a decrease in lipid saturation levels of LDs in hypoxic MIA PaCa2 cancer cells. This alteration in LD content is linked to reduced efficacy of treatments targeting cholesteryl ester formation. This study unveils important lipid metabolic changes in hypoxic cancer cells, providing insights that could lead to better treatment strategies for hypoxia-resistant cancer cells.
Collapse
Affiliation(s)
- Gil A. Gonzalez
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Ezinne U. Osuji
- College
of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, 201 S. University Street, West Lafayette, Indiana 47907, United States
| | - Natalie C. Fiur
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, 201 S. University Street, West Lafayette, Indiana 47907, United States
| | - Matthew G. Clark
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Seohee Ma
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Laura L. Lukov
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Chi Zhang
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, 201 S. University Street, West Lafayette, Indiana 47907, United States
- Purdue
Institute of Inflammation, Immunology, and Infectious Disease, 207 S. Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
16
|
Zuo B, Li X, Xu D, Zhao L, Yang Y, Luan Y, Zhang B. Targeting mitochondrial transfer: a new horizon in cardiovascular disease treatment. J Transl Med 2024; 22:1160. [PMID: 39741312 PMCID: PMC11687156 DOI: 10.1186/s12967-024-05979-x] [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: 09/29/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality among individuals with noncommunicable diseases worldwide. Obesity is associated with an increased risk of developing cardiovascular disease (CVD). Mitochondria are integral to the cardiovascular system, and it has been reported that mitochondrial transfer is associated with the pathogenesis of multiple CVDs and obesity. This review offers a comprehensive examination of the relevance of mitochondrial transfer to cardiovascular health and disease, emphasizing the critical functions of mitochondria in energy metabolism and signal transduction within the cardiovascular system. This highlights how disruptions in mitochondrial transfer contribute to various CVDs, such as myocardial infarction, cardiomyopathies, and hypertension. Additionally, we provide an overview of the molecular mechanisms governing mitochondrial transfer and its potential implications for CVD treatment. This finding underscores the therapeutic potential of mitochondrial transfer and addresses the various mechanisms and challenges in its implementation. By delving into mitochondrial transfer and its targeted modulation, this review aims to advance our understanding of cardiovascular disease treatment, presenting new insights and potential therapeutic strategies in this evolving field.
Collapse
Affiliation(s)
- Baile Zuo
- Molecular Immunology and Immunotherapy Laboratory, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
- Department of Clinical Laboratory, Heping Branch, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Dawei Xu
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Liping Zhao
- Department of Pathology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Bi Zhang
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| |
Collapse
|
17
|
Di Florio DN, Weigel GJ, Gorelov DJ, McCabe EJ, Beetler DJ, Shapiro KA, Bruno KA, Chekuri I, Jain A, Whelan ER, Salomon GR, Khatib S, Bonvie-Hill NE, Fliess JJ, Giresi PG, Hamilton C, Hartmoyer CJ, Balamurugan V, Darakjian AA, Edenfield BH, Kocsis SC, McLeod CJ, Cooper LT, Audet-Walsh É, Coronado MJ, Sin J, Fairweather D. Sex differences in mitochondrial gene expression during viral myocarditis. Biol Sex Differ 2024; 15:104. [PMID: 39696682 PMCID: PMC11657264 DOI: 10.1186/s13293-024-00678-0] [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: 12/06/2023] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Myocarditis is an inflammation of the heart muscle most often caused by viral infections. Sex differences in the immune response during myocarditis have been well described but upstream mechanisms in the heart that might influence sex differences in disease are not completely understood. METHODS Male and female BALB/c wild type mice received an intraperitoneal injection of heart-passaged coxsackievirus B3 (CVB3) or vehicle control. Bulk-tissue RNA-sequencing was conducted to better understand sex differences in CVB3 myocarditis. We performed enrichment analysis and functional validation to understand sex differences in the transcriptional landscape of myocarditis and identify factors that might drive sex differences in myocarditis. RESULTS As expected, the hearts of male and female mice with myocarditis were significantly enriched for pathways related to an innate and adaptive immune response compared to uninfected controls. Unique to this study, we found that males were enriched for inflammatory pathways and gene changes that suggested worse mitochondrial electron transport function while females were enriched for pathways related to mitochondrial homeostasis. Mitochondria isolated from the heart of males were confirmed to have worse mitochondrial respiration than females during myocarditis. Unbiased TRANSFAC analysis identified estrogen-related receptor alpha (ERRα) as a transcription factor that may mediate sex differences in mitochondrial function during myocarditis. Transcript and protein levels of ERRα were confirmed as elevated in females with myocarditis compared to males. Differential binding analysis from chromatin immunoprecipitation (ChIP) sequencing confirmed that ERRα bound highly to select predicted respiratory chain genes in females more than males during myocarditis. CONCLUSIONS Females with viral myocarditis regulate mitochondrial homeostasis by upregulating master regulators of mitochondrial transcription including ERRα.
Collapse
Affiliation(s)
- Damian N Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Gabriel J Weigel
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - David J Gorelov
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Elizabeth J McCabe
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Danielle J Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Katie A Shapiro
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Katelyn A Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Isha Chekuri
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Angita Jain
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Emily R Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Gary R Salomon
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Sami Khatib
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Jessica J Fliess
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Presley G Giresi
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Charwan Hamilton
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Ashley A Darakjian
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Brandy H Edenfield
- Department of Cancer Biology, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - S Christian Kocsis
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Division, CHU de Québec - Université Laval Research Center, Québec, QC, Canada
| | | | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, USA
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA.
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA.
- Department of Medicine, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
18
|
Wang N, Shang L, Liang Z, Feng M, Wang Y, Gao C, Luo J. Altered metabolic profiles of dermatomyositis with different myositis-specific autoantibodies associated with clinical phenotype. Front Immunol 2024; 15:1429010. [PMID: 39654882 PMCID: PMC11625817 DOI: 10.3389/fimmu.2024.1429010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Dermatomyositis (DM) is an idiopathic inflammatory myopathy. Because of clinical heterogeneity, the metabolite profile of DM patients with different myositis-specific autoantibodies (MSAs) remains elusive. This study aimed to explore the metabolomics characteristics of the serum in DM with different MSAs, low or high disease activity, and interstitial lung disease. Methods Untargeted metabolomics profiling was performed in the serum of a discovery cohort (n=96) and a validation cohort (n=40), consisting of DM patients with MSAs, low or high disease activity, and/or interstitial lung disease (DM-ILD) compared to age- and gender-matched healthy controls (HCs). Results The lipid profile in DM was found to be abnormal, especially dysregulated glycerophospholipid metabolism and fatty acid oxidation, which might affect the pathogenesis of DM by disrupting the balance of Th17 and Treg. We identified potential biomarkers of DM that can distinguish between low or high disease activity and reflect lung involvement. Two metabolite combinations including pro-leu, FA 14:0;O can distinguish high disease activity DM from low disease activity DM and HCs, and five including indole-3-lactic acid, dihydrosphingosine, SM 32:1;O2, NAE 17:1, and cholic acid can distinguish DM-ILD from DM without ILD (DM-nonILD). DM with different MSAs had unique metabolic characteristics, which can distinguish between MDA5+DM, Jo-1+DM, and TIF1-γ+DM, and from the antibody-negative groups. The sphingosine metabolism has been found to play an important role in MDA5+DM, which was associated with the occurrence of ILD. Discussion Altered metabolic profiles of dermatomyositis were associated with different myositisspecific autoantibodies, disease activity, and interstitial lung disease, which can help in the early diagnosis, prognosis, or selection of new therapeutic targets for DM.
Collapse
Affiliation(s)
- Nan Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lili Shang
- Shanxi Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Second Clinical Medical College, The Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhaojun Liang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Min Feng
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanlin Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chong Gao
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jing Luo
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
19
|
Cendrowski J, Wrobel M, Mazur M, Jary B, Maurya R, Wang S, Korostynski M, Dziewulska A, Rohm M, Kuropka P, Pudelko-Malik N, Mlynarz P, Dobrzyn A, Zeigerer A, Miaczynska M. NFκB and JNK pathways mediate metabolic adaptation upon ESCRT-I deficiency. Cell Mol Life Sci 2024; 81:458. [PMID: 39560723 DOI: 10.1007/s00018-024-05490-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/17/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024]
Abstract
Endosomal Sorting Complexes Required for Transport (ESCRTs) are crucial for delivering membrane receptors or intracellular organelles for lysosomal degradation which provides the cell with lysosome-derived nutrients. Yet, how ESCRT dysfunction affects cell metabolism remained elusive. To address this, we analyzed transcriptomes of cells lacking TSG101 or VPS28 proteins, components of ESCRT-I subcomplex. ESCRT-I deficiency reduced the expression of genes encoding enzymes involved in oxidation of fatty acids and amino acids, such as branched-chain amino acids, and increased the expression of genes encoding glycolytic enzymes. The changes in metabolic gene expression were associated with Warburg effect-like metabolic reprogramming that included intracellular accumulation of lipids, increased glucose/glutamine consumption and lactate production. Moreover, depletion of ESCRT-I components led to expansion of the ER and accumulation of small mitochondria, most of which retained proper potential and performed ATP-linked respiration. Mechanistically, the observed transcriptional reprogramming towards glycolysis in the absence of ESCRT-I occurred due to activation of the canonical NFκB and JNK signaling pathways and at least in part by perturbed lysosomal degradation. We propose that by activating the stress signaling pathways ESCRT-I deficiency leads to preferential usage of extracellular nutrients, like glucose and glutamine, for energy production instead of lysosome-derived nutrients, such as fatty acids and branched-chain amino acids.
Collapse
Affiliation(s)
- Jaroslaw Cendrowski
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Marta Wrobel
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Michal Mazur
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Bartosz Jary
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ranjana Maurya
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Surui Wang
- Institute for Diabetes and Cancer, Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, University Hospital, Heidelberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Anna Dziewulska
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, University Hospital, Heidelberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Patryk Kuropka
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Natalia Pudelko-Malik
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Piotr Mlynarz
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, University Hospital, Heidelberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marta Miaczynska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
| |
Collapse
|
20
|
Lumish HS, Sherrid MV, Janssen PML, Ferrari G, Hasegawa K, Castillero E, Adlestein E, Swistel DG, Topkara VK, Maurer MS, Reilly MP, Shimada YJ. Comprehensive Proteomic Profiling of Human Myocardium Reveals Signaling Pathways Dysregulated in Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2024; 84:1999-2011. [PMID: 39365226 PMCID: PMC11817648 DOI: 10.1016/j.jacc.2024.07.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiac disease. Signaling pathways that link genetic sequence variants to clinically overt HCM and progression to severe forms of HCM remain unknown. OBJECTIVES The purpose of this study was to identify signaling pathways that are differentially regulated in HCM, using proteomic profiling of human myocardium, confirmed with transcriptomic profiling. METHODS In this multicenter case-control study, myocardial samples were obtained from cases with HCM and control subjects with nonfailing hearts. Proteomic profiling of 7,289 proteins from myocardial samples was performed using the SomaScan assay (SomaLogic). Pathway analysis of differentially expressed proteins was performed, using a false discovery rate <0.05. Pathway analysis of proteins whose concentrations correlated with clinical indicators of severe HCM (eg, reduced left ventricular ejection fraction, atrial fibrillation, and ventricular tachyarrhythmias) was also executed. Confirmatory analysis of differentially expressed genes was performed using myocardial transcriptomic profiling. RESULTS The study included 99 HCM cases and 15 control subjects. Pathway analysis of differentially expressed proteins revealed dysregulation of the Ras-mitogen-activated protein kinase, ubiquitin-mediated proteolysis, angiogenesis-related (eg, hypoxia-inducible factor-1, vascular endothelial growth factor), and Hippo pathways. Pathways known to be dysregulated in HCM, including metabolic, inflammatory, and extracellular matrix pathways, were also dysregulated. Pathway analysis of proteins associated with clinical indicators of severe HCM and of differentially expressed genes supported these findings. CONCLUSIONS The present study represents the most comprehensive (>7,000 proteins) and largest-scale (n = 99 HCM cases) proteomic profiling of human HCM myocardium to date. Proteomic profiling and confirmatory transcriptomic profiling elucidate dysregulation of both newly recognized (eg, Ras-mitogen-activated protein kinase) and known pathways associated with pathogenesis and progression to severe forms of HCM.
Collapse
Affiliation(s)
- Heidi S Lumish
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Mark V Sherrid
- Leon Charney Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA
| | - Giovanni Ferrari
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA; Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Estibaliz Castillero
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Elizabeth Adlestein
- Leon Charney Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Daniel G Swistel
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, New York, USA
| | - Veli K Topkara
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA; Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| | - Yuichi J Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.
| |
Collapse
|
21
|
Zong N, Chowdhury S, Zhou S, Rajaganapathy S, Yu Y, Wang L, Dai Q, Li P, Liu X, Bielinski SJ, Chen J, Chen Y, Cerhan JR. Advancing Efficacy Prediction for EHR-based Emulated Trials in Repurposing Heart Failure Therapies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.05.25.23290531. [PMID: 37398384 PMCID: PMC10312819 DOI: 10.1101/2023.05.25.23290531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Introduction The High mortality rates associated with heart failure (HF) have propelled the strategy of drug repurposing, which seeks new therapeutic uses for existing, approved drugs to enhance the management of HF symptoms effectively. An emerging trend focuses on utilizing real-world data, like EHR, to mimic randomized controlled trials (RCTs) for evaluating treatment outcomes through what are known as emulated trials (ET). Nonetheless, the intricacies inherent in EHR data-comprising detailed patient histories in databases, the omission of certain biomarkers or specific diagnostic tests, and partial records of symptoms-introduce notable discrepancies between EHR data and the stringent standards of RCTs. This gap poses a substantial challenge in conducting an ET to accurately predict treatment efficacy. Objective The objective of this research is to predict the efficacy of drugs repurposed for HF in randomized trials by leveraging EHR in ET. Methods We proposed an ET framework to predict drug efficacy, integrating target prediction based on biomedical databases with statistical analysis using EHR data. Specifically, we developed a novel target prediction model that learns low-dimensional representations of drug molecules, protein sequences, and diverse biomedical associations from a knowledge graph. Additionally, we crafted strategies to improve the prediction by considering the interactions between HF drugs and biological factors in the context of HF prognostic markers. Results Our validation of the drug-target prediction model against the BETA benchmark demonstrated superior performance, with an average AUCROC of 97.7%, PRAUC of 97.4%, F1 score of 93.1%, and a General Score of 96.1%, surpassing existing baseline algorithms. Further analysis of our ET framework on identifying 17 repurposed drugs-derived from 266 phase 3 HF RCTs-using data from 59,000 patients at the Mayo Clinic highlighted the framework's remarkable predictive accuracy. This analysis took into account various factors such as biological variables (e.g., gender, age, ethnicity), HF medications (e.g., ACE inhibitors, Beta-blockers, ARBs, Loop Diuretics), types of HF (HFpEF and HFrEF), confounders, and prognostic markers (e.g., NT-proBNP, bUn, creatinine, and hemoglobin). The ET framework significantly improved the accuracy compared to the baseline efficacy analysis that utilized EHR data. Notably, the best results were improved in AUC-ROC from 75.71% to 93.57% and in PRAUC from 78.66% to 90.34%, compared to the baseline models. Conclusion Our study presents an ET framework that significantly enhances drug efficacy emulation by integrating EHR-based analysis with target prediction. We demonstrated substantial success in predicting the efficacy of 17 HF drugs repurposed for phase 3 RCTs, showcasing the framework's potential in advancing HF treatment strategies.
Collapse
Affiliation(s)
- Nansu Zong
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Shaika Chowdhury
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Shibo Zhou
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Sivaraman Rajaganapathy
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Yue Yu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Qiying Dai
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Pengyang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, VA, USA
| | - Xiaoke Liu
- Division of Community Cardiology, Department of Cardiovascular Medicine, La Crosse, Wisconsin, WI, USA
| | | | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Yongbin Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - James R. Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
22
|
Deng K, Hua Y, Gao Y, Zheng H, Jiang Y, Wang Y, Gao C, Ren T, Zhu Y. Thermosensitive Hydrogel with Programmable, Self-Regulated HIF-1α Stabilizer Release for Myocardial Infarction Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408013. [PMID: 39308185 DOI: 10.1002/advs.202408013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/27/2024] [Indexed: 11/22/2024]
Abstract
HIF-1α (hypoxia induced factor-1α), a vital protective signal against hypoxia, has a short lifetime after myocardial infarction (MI). Increasing HIF-1α stability by inhibiting its hydroxylation with prolyl hydroxylases inhibitors such as DPCA (1,4-dihydrophenonthrolin-4-one-3-carboxylic acid) presents positive results. However, the optimal inhibitor administration profile for MI treatment is still unexplored. Here, injectable, thermosensitive hydrogels with programmable DPCA release are designed and synthesized. Hydrogel degradation and slow DPCA release are coupled to form a feedback loop by attaching pendant DPCA to polymer backbone, which serve as additional crosslinking points through π-π and hydrophobic interactions. Pendant carboxyl groups are added to the copolymer to accelerate DPCA release. Burst release in the acute phase for myocardial protection and extended near zero-order release across the inflammatory and fibrotic phases with different rates are achieved. All DPCA-releasing hydrogels upregulate HIF-1α, decrease apoptosis, promote angiogenesis, and stimulate cardiomyocyte proliferation, leading to preserved cardiac function and ventricular geometry. Faster hydrogel degradation induced by faster DPCA release results in a HIF-1α expression eight times of healthy control and better therapeutic effect in MI treatment. This research demonstrates the value of precise regulation of HIF-1α expression in treating MI and other relevant diseases and provides an implantable device-based modulation strategy.
Collapse
Affiliation(s)
- Kaicheng Deng
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuyan Hua
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ying Gao
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Houwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yangzi Jiang
- School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, Hong Kong, 999077, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, Hong Kong, 999077, China
| | - Yaping Wang
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Changyou Gao
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tanchen Ren
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Yang Zhu
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| |
Collapse
|
23
|
Ravindran S, Rau CD. The multifaceted role of mitochondria in cardiac function: insights and approaches. Cell Commun Signal 2024; 22:525. [PMID: 39472951 PMCID: PMC11523909 DOI: 10.1186/s12964-024-01899-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
Cardiovascular disease (CVD) remains a global economic burden even in the 21st century with 85% of deaths resulting from heart attacks. Despite efforts in reducing the risk factors, and enhancing pharmacotherapeutic strategies, challenges persist in early identification of disease progression and functional recovery of damaged hearts. Targeting mitochondrial dysfunction, a key player in the pathogenesis of CVD has been less successful due to its role in other coexisting diseases. Additionally, it is the only organelle with an agathokakological function that is a remedy and a poison for the cell. In this review, we describe the origins of cardiac mitochondria and the role of heteroplasmy and mitochondrial subpopulations namely the interfibrillar, subsarcolemmal, perinuclear, and intranuclear mitochondria in maintaining cardiac function and in disease-associated remodeling. The cumulative evidence of mitochondrial retrograde communication with the nucleus is addressed, highlighting the need to study the genotype-phenotype relationships of specific organelle functions with CVD by using approaches like genome-wide association study (GWAS). Finally, we discuss the practicality of computational methods combined with single-cell sequencing technologies to address the challenges of genetic screening in the identification of heteroplasmy and contributory genes towards CVD.
Collapse
Affiliation(s)
- Sriram Ravindran
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA
| | - Christoph D Rau
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA.
| |
Collapse
|
24
|
Xu J, Chen WJ, Hu HB, Xie ZW, Zhang DG, Zhao J, Xiang J, Wei QY, Tidwell T, Girard O, Ma FH, Li ZW, Ren YM. A global view on quantitative proteomic and metabolic analysis of rat livers under different hypoxia protocols. Heliyon 2024; 10:e37791. [PMID: 39381102 PMCID: PMC11456861 DOI: 10.1016/j.heliyon.2024.e37791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 08/01/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Hypobaric hypoxia causes altitude sickness and significantly affects human health. As of now, focusing on rats different proteomic and metabolic changes exposed to different hypoxic times at extreme altitude is blank. Our study integrated in vivo experiments with tandem mass tag (TMT)- and gas chromatography time-of-flight (GC-TOF)-based proteomic and metabolomic assessments, respectively. Male Sprague-Dawley rats were exposed to long-term constant hypoxia for 40 days or short-term constant hypoxia for three days, and their responses were compared with those of a normal control group. Post-hypoxia, serum marker assays related to lipid metabolism revealed significant increases in the levels of low-density lipoprotein (LDL), triglycerides (TG), and total cholesterol (TC) in the liver. In contrast, high-density lipoprotein (HDL) levels were upregulated in the long-term constant hypoxia cohorts and were significantly reduced in the short-term constant hypoxia cohorts. Furthermore, metabolic pathway analysis indicated that glycerolipid and glycerophospholipid metabolisms were the most significantly affected pathways in long-term hypoxia group. Subsequently, RT-qPCR analyses were performed to corroborate the key regulatory elements, including macrophage galactose-type lectin (MGL) and Fatty Acid Desaturase 2 (FADS2). The results of this study provide new information for understanding the effects of different hypobaric hypoxia exposure protocols on protein expression and metabolism in low-altitude animals.
Collapse
Affiliation(s)
- Jin Xu
- Qinghai University, Xining, 810001, China
| | | | | | | | | | - Jia Zhao
- Qinghai University, Xining, 810001, China
| | - Jing Xiang
- Qinghai University, Xining, 810001, China
| | - Qi-yu Wei
- Qinghai University, Xining, 810001, China
| | - Tawni Tidwell
- Center for Healthy Minds, University of Wisconsin-Madison, 625 Washington Ave, Madison, WI, 53711, USA
| | - Olivier Girard
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia, Crawley, Western Australia, Australia
| | - Fu-hai Ma
- Qinghai Institute of Sports Science, Xi Ning, China
| | | | | |
Collapse
|
25
|
Findley TO, Palei AC, Cho KS, Zhao Z, Shi C, Mahajan G, Corno AF, Salazar J, McCullough L. Sex differences in metabolic adaptation in infants with cyanotic congenital heart disease. Pediatr Res 2024; 96:1201-1209. [PMID: 38839995 PMCID: PMC11524789 DOI: 10.1038/s41390-024-03291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/23/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Female infants with congenital heart disease (CHD) face significantly higher postoperative mortality rates after adjusting for cardiac complexity. Sex differences in metabolic adaptation to cardiac stressors may be an early contributor to cardiac dysfunction. In adult diseases, hypoxic/ischemic cardiomyocytes undergo a cardioprotective metabolic shift from oxidative phosphorylation to glycolysis which appears to be regulated in a sexually dimorphic manner. We hypothesize sex differences in cardiac metabolism are present in cyanotic CHD and detectable as early as the infant period. METHODS RNA sequencing was performed on blood samples (cyanotic CHD cases, n = 11; controls, n = 11) and analyzed using gene set enrichment analysis (GSEA). Global plasma metabolite profiling (UPLC-MS/MS) was performed using a larger representative cohort (cyanotic CHD, n = 27; non-cyanotic CHD, n = 11; unaffected controls, n = 12). RESULTS Hallmark gene sets in glycolysis, fatty acid metabolism, and oxidative phosphorylation were significantly enriched in cyanotic CHD females compared to male counterparts, which was consistent with metabolomic differences between sexes. Minimal sex differences in metabolic pathways were observed in normoxic patients (both controls and non-cyanotic CHD cases). CONCLUSION These observations suggest underlying differences in metabolic adaptation to chronic hypoxia between males and females with cyanotic CHD. IMPACT Children with cyanotic CHD exhibit sex differences in utilization of glycolysis vs. fatty acid oxidation pathways to meet the high-energy demands of the heart in the neonatal period. Transcriptomic and metabolomic results suggest that under hypoxic conditions, males and females undergo metabolic shifts that are sexually dimorphic. These sex differences were not observed in neonates in normoxic conditions (i.e., non-cyanotic CHD and unaffected controls). The involved metabolic pathways are similar to those observed in advanced heart failure, suggesting metabolic adaptations beginning in the neonatal period may contribute to sex differences in infant survival.
Collapse
Affiliation(s)
- Tina O Findley
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, USA.
| | - Ana Carolina Palei
- Department of Surgery, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kyung Serk Cho
- Center for Precision Health, School of Biomedical Informatics at the University of Texas Health Science Center Houston, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics at the University of Texas Health Science Center Houston, Houston, TX, USA
- Human Genetics Center, School of Public Health at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Caleb Shi
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Gouri Mahajan
- Department of Pharmacology and Toxicology/Biobank, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Jorge Salazar
- Children's Heart Institute, McGovern Medical School at the University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Louise McCullough
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
26
|
Zhang H, Zhang Y, Zhang J, Jia D. Exercise Alleviates Cardiovascular Diseases by Improving Mitochondrial Homeostasis. J Am Heart Assoc 2024; 13:e036555. [PMID: 39291488 DOI: 10.1161/jaha.124.036555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Engaging in regular exercise and physical activity contributes to delaying the onset of cardiovascular diseases (CVDs). However, the physiological mechanisms underlying the benefits of regular exercise or physical activity in CVDs remain unclear. The disruption of mitochondrial homeostasis is implicated in the pathological process of CVDs. Exercise training effectively delays the onset and progression of CVDs by significantly ameliorating the disruption of mitochondrial homeostasis. This includes improving mitochondrial biogenesis, increasing mitochondrial fusion, decreasing mitochondrial fission, promoting mitophagy, and mitigating mitochondrial morphology and function. This review provides a comprehensive overview of the benefits of physical exercise in the context of CVDs, establishing a connection between the disruption of mitochondrial homeostasis and the onset of these conditions. Through a detailed examination of the underlying molecular mechanisms within mitochondria, the study illuminates how exercise can provide innovative perspectives for future therapies for CVDs.
Collapse
Affiliation(s)
- Huijie Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Yuxuan Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Jiaqiao Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Dandan Jia
- School of Exercise and health Shanghai University of Sport Shanghai China
| |
Collapse
|
27
|
Jonker PB, Muir A. Metabolic ripple effects - deciphering how lipid metabolism in cancer interfaces with the tumor microenvironment. Dis Model Mech 2024; 17:dmm050814. [PMID: 39284708 PMCID: PMC11423921 DOI: 10.1242/dmm.050814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
Cancer cells require a constant supply of lipids. Lipids are a diverse class of hydrophobic molecules that are essential for cellular homeostasis, growth and survival, and energy production. How tumors acquire lipids is under intensive investigation, as these mechanisms could provide attractive therapeutic targets for cancer. Cellular lipid metabolism is tightly regulated and responsive to environmental stimuli. Thus, lipid metabolism in cancer is heavily influenced by the tumor microenvironment. In this Review, we outline the mechanisms by which the tumor microenvironment determines the metabolic pathways used by tumors to acquire lipids. We also discuss emerging literature that reveals that lipid availability in the tumor microenvironment influences many metabolic pathways in cancers, including those not traditionally associated with lipid biology. Thus, metabolic changes instigated by the tumor microenvironment have 'ripple' effects throughout the densely interconnected metabolic network of cancer cells. Given the interconnectedness of tumor metabolism, we also discuss new tools and approaches to identify the lipid metabolic requirements of cancer cells in the tumor microenvironment and characterize how these requirements influence other aspects of tumor metabolism.
Collapse
Affiliation(s)
- Patrick B Jonker
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
28
|
Mialet-Perez J, Belaidi E. Interplay between hypoxia inducible Factor-1 and mitochondria in cardiac diseases. Free Radic Biol Med 2024; 221:13-22. [PMID: 38697490 DOI: 10.1016/j.freeradbiomed.2024.04.239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Ischemic heart diseases and cardiomyopathies are characterized by hypoxia, energy starvation and mitochondrial dysfunction. HIF-1 acts as a cellular oxygen sensor, tuning the balance of metabolic and oxidative stress pathways to provide ATP and sustain cell survival. Acting on mitochondria, HIF-1 regulates different processes such as energy substrate utilization, oxidative phosphorylation and mitochondrial dynamics. In turn, mitochondrial homeostasis modifications impact HIF-1 activity. This underlies that HIF-1 and mitochondria are tightly interconnected to maintain cell homeostasis. Despite many evidences linking HIF-1 and mitochondria, the mechanistic insights are far from being understood, particularly in the context of cardiac diseases. Here, we explore the current understanding of how HIF-1, reactive oxygen species and cell metabolism are interconnected, with a specific focus on mitochondrial function and dynamics. We also discuss the divergent roles of HIF in acute and chronic cardiac diseases in order to highlight that HIF-1, mitochondria and oxidative stress interaction deserves to be deeply investigated. While the strategies aiming at stabilizing HIF-1 have provided beneficial effects in acute ischemic injury, some deleterious effects were observed during prolonged HIF-1 activation. Thus, deciphering the link between HIF-1 and mitochondria will help to optimize HIF-1 modulation and provide new therapeutic perspectives for the treatment of cardiovascular pathologies.
Collapse
Affiliation(s)
- Jeanne Mialet-Perez
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, France
| | - Elise Belaidi
- Univ. Lyon 1, Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, LBTI UMR 5305, 69367, Lyon, France.
| |
Collapse
|
29
|
Hu X, Jiang D, Zhang Z, An Z. PPAR-α regulates metabolic remodelling and participates in myocardial fibrosis in patients with atrial fibrillation of rheumatic heart disease. Arch Med Sci 2024; 20:1461-1471. [PMID: 39649284 PMCID: PMC11623188 DOI: 10.5114/aoms/181134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/12/2024] [Indexed: 12/10/2024] Open
Abstract
Introduction This study will explore the correlation of peroxisome proliferator activated receptor-α (PPAR-α) regulation of metabolic remodelling in the myocardial fibrosis of atrial fibrillation (AF) in rheumatic heart disease. Material and methods The left atrial appendage tissues were evaluated by Masson staining for fibrosis degree, and Western Blot was used to detect the expression of proteins related to glucose metabolism disorder, lipid metabolism abnormality, and mitochondrial dysfunction. The myocardial fibroblasts were established by stimulation with ANG II, and the PPAR-α agonist GW7647 was administered. The changes of phenotype transformation of myocardial fibroblasts were detected by cellular immunofluorescence, the secretion level of supernatant collagen was detected by ELISA. Finally, the correlation between PPAR-α protein expression and myocardial fibrosis was analysed and a conclusion was drawn. Results Masson staining showed that the degree of myocardial fibrosis in patients with AF was significantly increased; WB analysis showed that there were statistically significant differences in protein expression related to glucose metabolism disorder, lipid metabolism abnormality, and mitochondrial dysfunction. There was a correlation between PPAR-α protein expression and myocardial fibrosis (r = -0.5322, p < 0.0001). After stimulation with PPAR-α agonist GW7647, the phenotypic differentiation of myocardial fibro-blasts into myofibroblasts was inhibited. The protein expression related to mitochondrial dysfunction was statistically different. Conclusions This study found that there is a negative correlation between the expression of PPAR-α protein and myocardial fibrosis in rheumatic heart disease AF, which plays a protective role. PPAR-α may participate in the pathogenesis of myocardial fibrosis in rheumatic heart disease AF by regulating glucose metabolism, lipid metabolism, and mitochondrial function.
Collapse
Affiliation(s)
- Xiaoying Hu
- Department of Laboratory Medicine, Zigong Fourth People’s Hospital, Zigong, China
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Daisong Jiang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenmei An
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Zuo B, Fan X, Xu D, Zhao L, Zhang B, Li X. Deciphering the mitochondria-inflammation axis: Insights and therapeutic strategies for heart failure. Int Immunopharmacol 2024; 139:112697. [PMID: 39024750 DOI: 10.1016/j.intimp.2024.112697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/20/2024]
Abstract
Heart failure (HF) is a clinical syndrome resulting from left ventricular systolic and diastolic dysfunction, leading to significant morbidity and mortality worldwide. Despite improvements in medical treatment, the prognosis of HF patients remains unsatisfactory, with high rehospitalization rates and substantial economic burdens. The heart, a high-energy-consuming organ, relies heavily on ATP production through oxidative phosphorylation in mitochondria. Mitochondrial dysfunction, characterized by impaired energy production, oxidative stress, and disrupted calcium homeostasis, plays a crucial role in HF pathogenesis. Additionally, inflammation contributes significantly to HF progression, with elevated levels of circulating inflammatory cytokines observed in patients. The interplay between mitochondrial dysfunction and inflammation involves shared risk factors, signaling pathways, and potential therapeutic targets. This review comprehensively explores the mechanisms linking mitochondrial dysfunction and inflammation in HF, including the roles of mitochondrial reactive oxygen species (ROS), calcium dysregulation, and mitochondrial DNA (mtDNA) release in triggering inflammatory responses. Understanding these complex interactions offers insights into novel therapeutic approaches for improving mitochondrial function and relieving oxidative stress and inflammation. Targeted interventions that address the mitochondria-inflammation axis hold promise for enhancing cardiac function and outcomes in HF patients.
Collapse
Affiliation(s)
- Baile Zuo
- Molecular Immunology and Immunotherapy Laboratory, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiu Fan
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Dawei Xu
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Liping Zhao
- Department of Pathology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Bi Zhang
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China; Department of Clinical Laboratory, Heping Branch, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| |
Collapse
|
31
|
Chen X, Qian W, Zhang Y, Zhao P, Lin X, Yang S, Zhuge Q, Ni H. Ginsenoside CK cooperates with bone mesenchymal stem cells to enhance angiogenesis post-stroke via GLUT1 and HIF-1α/VEGF pathway. Phytother Res 2024. [PMID: 38990183 DOI: 10.1002/ptr.8235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 07/12/2024]
Abstract
The transplantation of bone marrow mesenchymal stem cells (MSCs) in stroke is hindered by the restricted rates of survival and differentiation. Ginsenoside compound K (CK), is reported to have a neuroprotective effect and regulate energy metabolism. We applied CK to investigate if CK could promote the survival of MSCs and differentiation into brain microvascular endothelial-like cells (BMECs), thereby alleviating stroke symptoms. Therefore, transwell and middle cerebral artery occlusion (MCAO) models were used to mimic oxygen and glucose deprivation (OGD) in vitro and in vivo, respectively. Our results demonstrated that CK had a good affinity for GLUT1, which increased the expression of GLUT1 and the production of ATP, facilitated the proliferation and migration of MSCs, and activated the HIF-1α/VEGF signaling pathway to promote MSC differentiation. Moreover, CK cooperated with MSCs to protect BMECs, promote angiogenesis and vascular density, enhance neuronal and astrocytic proliferation, thereby reducing infarct volume and consequently improving neurobehavioral outcomes. These results suggest that the synergistic effects of CK and MSCs could potentially be a promising strategy for stroke.
Collapse
Affiliation(s)
- Xijun Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenqi Qian
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peiqi Zhao
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangxiang Lin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Su Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haoqi Ni
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
32
|
Hinton A, Claypool SM, Neikirk K, Senoo N, Wanjalla CN, Kirabo A, Williams CR. Mitochondrial Structure and Function in Human Heart Failure. Circ Res 2024; 135:372-396. [PMID: 38963864 PMCID: PMC11225798 DOI: 10.1161/circresaha.124.323800] [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] [Indexed: 07/06/2024]
Abstract
Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Steven M. Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Nanami Senoo
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH (C.R.W.)
| |
Collapse
|
33
|
Rubio-Tomás T, Soler-Botija C, Martínez-Estrada O, Villena JA. Transcriptional control of cardiac energy metabolism in health and disease: Lessons from animal models. Biochem Pharmacol 2024; 224:116185. [PMID: 38561091 DOI: 10.1016/j.bcp.2024.116185] [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: 12/11/2023] [Revised: 02/27/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Cardiac ATP production is tightly regulated in order to satisfy the evolving energetic requirements imposed by different cues during health and pathological conditions. In order to sustain high ATP production rates, cardiac cells are endowed with a vast mitochondrial network that is essentially acquired during the perinatal period. Nevertheless, adult cardiac cells also adapt their mitochondrial mass and oxidative function to changes in energy demand and substrate availability by fine-tuning the pathways and mitochondrial machinery involved in energy production. The reliance of cardiac cells on mitochondrial metabolism makes them particularly sensitive to alterations in proper mitochondrial function, so that deficiency in energy production underlies or precipitates the development of heart diseases. Mitochondrial biogenesis is a complex process fundamentally controlled at the transcriptional level by a network of transcription factors and co-regulators, sometimes with partially redundant functions, that ensure adequate energy supply to the working heart. Novel uncovered regulators, such as RIP140, PERM1, MED1 or BRD4 have been recently shown to modulate or facilitate the transcriptional activity of the PGC-1s/ERRs/PPARs regulatory axis, allowing cardiomyocytes to adapt to a variety of physiological or pathological situations requiring different energy provision. In this review, we summarize the current knowledge on the mechanisms that regulate cardiac mitochondrial biogenesis, highlighting the recent discoveries of new transcriptional regulators and describing the experimental models that have provided solid evidence of the relevant contribution of these factors to cardiac function in health and disease.
Collapse
Affiliation(s)
- Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion GR-70013, Crete, Greece
| | - Carolina Soler-Botija
- ICREC (Heart Failure and Cardiac Regeneration) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBER on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Josep A Villena
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain.
| |
Collapse
|
34
|
Lee C, Liang LW, Hasegawa K, Maurer MS, Tower-Rader A, Fifer MA, Reilly M, Shimada YJ. Signaling Pathways Associated With Prior Cardiovascular Events in Hypertrophic Cardiomyopathy. J Card Fail 2024; 30:462-472. [PMID: 37562580 PMCID: PMC10853480 DOI: 10.1016/j.cardfail.2023.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiomyopathy. A subset of patients experience major adverse cardiovascular events (MACEs), including arrhythmias, strokes and heart failure. However, the molecular mechanisms underlying MACEs in HCM are still not well understood. Therefore, we conducted a multicenter case-control study of patients with HCM, comparing those with and without prior histories of MACEs to identify dysregulated signaling pathways through plasma proteomics profiling. METHODS We performed plasma proteomics profiling of 4986 proteins. We developed a proteomics-based discrimination model in patients enrolled at 1 institution (training set) and externally validated the model in patients enrolled at another institution (test set). We performed pathway analysis of proteins dysregulated in patients with prior MACEs. RESULTS A total of 402 patients were included, with 278 in the training set and 124 in the test set. In this cohort, 257 (64%) patients had prior MACEs (172 in the training set and 85 in the test set). Using the proteomics-based model from the training set, the area under the receiver operating characteristic curve was 0.82 (95% confidence interval, 0.75-0.90) in the test set. Patients with prior MACEs demonstrated dysregulation in pathways known to be associated with MACEs (eg, TGF-β) and novel pathways (eg, Ras-MAPK and associated pathways). CONCLUSIONS In this multicenter study of 402 patients with HCM, we identified both known and novel pathways dysregulated in a subset of patients with more advanced disease.
Collapse
Affiliation(s)
- Charlotte Lee
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Lusha W Liang
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Albree Tower-Rader
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Michael A Fifer
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Muredach Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY; Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY
| | - Yuichi J Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
35
|
Shen C, Ding X, Ruan J, Ruan F, Hu W, Huang J, He C, Yu Y, Zuo Z. Black phosphorus quantum dots induce myocardial inflammatory responses and metabolic disorders in mice. J Environ Sci (China) 2024; 137:53-64. [PMID: 37980037 DOI: 10.1016/j.jes.2023.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 11/20/2023]
Abstract
As an ultrasmall derivative of black phosphorus (BP) sheets, BP quantum dots (BP-QDs) have been effectively used in many fields. Currently, information on the cardiotoxicity induced by BP-QDs remains limited. We aimed to evaluate BP-QD-induced cardiac toxicity in mice. Histopathological examination of heart tissue sections was performed. Transcriptome sequencing, real-time quantitative PCR (RT‒qPCR), western blotting, and enzyme-linked immunosorbent assay (ELISA) assays were used to detect the mRNA and/or protein expression of proinflammatory cytokines, nuclear factor kappa B (NF-κB), phosphatidylinositol 3 kinase-protein kinase B (PI3K-AKT), peroxisome proliferator-activated receptor gamma (PPARγ), and glucose/lipid metabolism pathway-related genes. We found that heart weight and heart/body weight index (HBI) were significantly reduced in mice after intragastric administration of 0.1 or 1 mg/kg BP-QDs for 28 days. In addition, obvious inflammatory cell infiltration and increased cardiomyocyte diameter were observed in the BP-QD-treated groups. Altered expression of proinflammatory cytokines and genes related to the NF-κB signaling pathway further confirmed that BP-QD exposure induced inflammatory responses. In addition, BP-QD treatment also affected the PI3K-AKT, PPARγ, thermogenesis, oxidative phosphorylation, and cardiac muscle contraction signaling pathways. The expression of genes related to glucose/lipid metabolism signaling pathways was dramatically affected by BP-QD exposure, and the effect was primarily mediated by the PPAR signaling pathway. Our study provides new insights into the toxicity of BP-QDs to human health.
Collapse
Affiliation(s)
- Chao Shen
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xiaoyan Ding
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jinpeng Ruan
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Fengkai Ruan
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Weiping Hu
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jiyi Huang
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Chengyong He
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yi Yu
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Zhenghong Zuo
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
36
|
Froese N, Szaroszyk M, Galuppo P, Visker JR, Werlein C, Korf‐Klingebiel M, Berliner D, Reboll MR, Hamouche R, Gegel S, Wang Y, Hofmann W, Tang M, Geffers R, Wende AR, Kühnel MP, Jonigk DD, Hansmann G, Wollert KC, Abel ED, Drakos SG, Bauersachs J, Riehle C. Hypoxia Attenuates Pressure Overload-Induced Heart Failure. J Am Heart Assoc 2024; 13:e033553. [PMID: 38293923 PMCID: PMC11056135 DOI: 10.1161/jaha.123.033553] [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: 11/14/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Alveolar hypoxia is protective in the context of cardiovascular and ischemic heart disease; however, the underlying mechanisms are incompletely understood. The present study sought to test the hypothesis that hypoxia is cardioprotective in left ventricular pressure overload (LVPO)-induced heart failure. We furthermore aimed to test that overlapping mechanisms promote cardiac recovery in heart failure patients following left ventricular assist device-mediated mechanical unloading and circulatory support. METHODS AND RESULTS We established a novel murine model of combined chronic alveolar hypoxia and LVPO following transverse aortic constriction (HxTAC). The HxTAC model is resistant to cardiac hypertrophy and the development of heart failure. The cardioprotective mechanisms identified in our HxTAC model include increased activation of HIF (hypoxia-inducible factor)-1α-mediated angiogenesis, attenuated induction of genes associated with pathological remodeling, and preserved metabolic gene expression as identified by RNA sequencing. Furthermore, LVPO decreased Tbx5 and increased Hsd11b1 mRNA expression under normoxic conditions, which was attenuated under hypoxic conditions and may induce additional hypoxia-mediated cardioprotective effects. Analysis of samples from patients with advanced heart failure that demonstrated left ventricular assist device-mediated myocardial recovery revealed a similar expression pattern for TBX5 and HSD11B1 as observed in HxTAC hearts. CONCLUSIONS Hypoxia attenuates LVPO-induced heart failure. Cardioprotective pathways identified in the HxTAC model might also contribute to cardiac recovery following left ventricular assist device support. These data highlight the potential of our novel HxTAC model to identify hypoxia-mediated cardioprotective mechanisms and therapeutic targets that attenuate LVPO-induced heart failure and mediate cardiac recovery following mechanical circulatory support.
Collapse
Affiliation(s)
- Natali Froese
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | | | - Paolo Galuppo
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - Joseph R. Visker
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI) and Division of Cardiovascular MedicineUniversity of Utah School of MedicineSalt Lake CityUTUSA
| | | | | | - Dominik Berliner
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - Marc R. Reboll
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - Rana Hamouche
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI) and Division of Cardiovascular MedicineUniversity of Utah School of MedicineSalt Lake CityUTUSA
| | - Simona Gegel
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - Yong Wang
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - Winfried Hofmann
- Department of Human GeneticsHannover Medical SchoolHannoverGermany
| | - Ming Tang
- Department of Human GeneticsHannover Medical SchoolHannoverGermany
- L3S Research CenterLeibniz UniversityHannoverGermany
| | - Robert Geffers
- Helmholtz Center for Infection ResearchResearch Group Genome AnalyticsBraunschweigGermany
| | - Adam R. Wende
- Division of Molecular and Cellular Pathology, Department of PathologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - Mark P. Kühnel
- Institute of PathologyHannover Medical SchoolHannoverGermany
- Biomedical Research in End‐stage and Obstructive Lung Disease Hannover (BREATH)German Lung Research Center (DZL)HannoverGermany
| | - Danny D. Jonigk
- Institute of PathologyHannover Medical SchoolHannoverGermany
- Biomedical Research in End‐stage and Obstructive Lung Disease Hannover (BREATH)German Lung Research Center (DZL)HannoverGermany
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical CareHannover Medical SchoolHannoverGermany
- Department of Pediatric CardiologyUniversity Medical Center Erlangen, Friedrich‐Alexander University Erlangen‐NürnbergErlangenGermany
| | - Kai C. Wollert
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - E. Dale Abel
- Department of MedicineDavid Geffen School of Medicine and UCLA HealthLos AngelesCAUSA
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI) and Division of Cardiovascular MedicineUniversity of Utah School of MedicineSalt Lake CityUTUSA
| | - Johann Bauersachs
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - Christian Riehle
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| |
Collapse
|
37
|
Maestri A, Garagnani P, Pedrelli M, Hagberg CE, Parini P, Ehrenborg E. Lipid droplets, autophagy, and ageing: A cell-specific tale. Ageing Res Rev 2024; 94:102194. [PMID: 38218464 DOI: 10.1016/j.arr.2024.102194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Lipid droplets are the essential organelle for storing lipids in a cell. Within the variety of the human body, different cells store, utilize and release lipids in different ways, depending on their intrinsic function. However, these differences are not well characterized and, especially in the context of ageing, represent a key factor for cardiometabolic diseases. Whole body lipid homeostasis is a central interest in the field of cardiometabolic diseases. In this review we characterize lipid droplets and their utilization via autophagy and describe their diverse fate in three cells types central in cardiometabolic dysfunctions: adipocytes, hepatocytes, and macrophages.
Collapse
Affiliation(s)
- Alice Maestri
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Matteo Pedrelli
- Cardio Metabolic Unit, Department of Laboratory Medicine, and Department of Medicine (Huddinge), Karolinska Institutet, Stockholm, Sweden; Medicine Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Parini
- Cardio Metabolic Unit, Department of Laboratory Medicine, and Department of Medicine (Huddinge), Karolinska Institutet, Stockholm, Sweden; Medicine Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Ewa Ehrenborg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
38
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [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: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
39
|
Dörmann N, Hammer E, Struckmann K, Rüdebusch J, Bartels K, Wenzel K, Schulz J, Gross S, Schwanz S, Martin E, Fielitz B, Pablo Tortola C, Hahn A, Benkner A, Völker U, Felix SB, Fielitz J. Metabolic remodeling in cardiac hypertrophy and heart failure with reduced ejection fraction occurs independent of transcription factor EB in mice. Front Cardiovasc Med 2024; 10:1323760. [PMID: 38259303 PMCID: PMC10800928 DOI: 10.3389/fcvm.2023.1323760] [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: 10/18/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Background A metabolic shift from fatty acid (FAO) to glucose oxidation (GO) occurs during cardiac hypertrophy (LVH) and heart failure with reduced ejection fraction (HFrEF), which is mediated by PGC-1α and PPARα. While the transcription factor EB (TFEB) regulates the expression of both PPARGC1A/PGC-1α and PPARA/PPARα, its contribution to metabolic remodeling is uncertain. Methods Luciferase assays were performed to verify that TFEB regulates PPARGC1A expression. Cardiomyocyte-specific Tfeb knockout (cKO) and wildtype (WT) male mice were subjected to 27G transverse aortic constriction or sham surgery for 21 and 56 days, respectively, to induce LVH and HFrEF. Echocardiographic, morphological, and histological analyses were performed. Changes in markers of cardiac stress and remodeling, metabolic shift and oxidative phosphorylation were investigated by Western blot analyses, mass spectrometry, qRT-PCR, and citrate synthase and complex II activity measurements. Results Luciferase assays revealed that TFEB increases PPARGC1A/PGC-1α expression, which was inhibited by class IIa histone deacetylases and derepressed by protein kinase D. At baseline, cKO mice exhibited a reduced cardiac function, elevated stress markers and a decrease in FAO and GO gene expression compared to WT mice. LVH resulted in increased cardiac remodeling and a decreased expression of FAO and GO genes, but a comparable decline in cardiac function in cKO compared to WT mice. In HFrEF, cKO mice showed an improved cardiac function, lower heart weights, smaller myocytes and a reduction in cardiac remodeling compared to WT mice. Proteomic analysis revealed a comparable decrease in FAO- and increase in GO-related proteins in both genotypes. A significant reduction in mitochondrial quality control genes and a decreased citrate synthase and complex II activities was observed in hearts of WT but not cKO HFrEF mice. Conclusions TFEB affects the baseline expression of metabolic and mitochondrial quality control genes in the heart, but has only minor effects on the metabolic shift in LVH and HFrEF in mice. Deletion of TFEB plays a protective role in HFrEF but does not affect the course of LVH. Further studies are needed to elucidate if TFEB affects the metabolic flux in stressed cardiomyocytes.
Collapse
Affiliation(s)
- Niklas Dörmann
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Elke Hammer
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Karlotta Struckmann
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Julia Rüdebusch
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Kirsten Bartels
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Kristin Wenzel
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Julia Schulz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Stefan Gross
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Stefan Schwanz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Elisa Martin
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Britta Fielitz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Cristina Pablo Tortola
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Hahn
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Benkner
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B. Felix
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Jens Fielitz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
40
|
Carvalho RA. The glycolytic pathway to heart failure. GLYCOLYSIS 2024:235-266. [DOI: 10.1016/b978-0-323-91704-9.00010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
41
|
Martin-Puig S, Menendez-Montes I. Cardiac Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:365-396. [PMID: 38884721 DOI: 10.1007/978-3-031-44087-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The heart is composed of a heterogeneous mixture of cellular components perfectly intermingled and able to integrate common environmental signals to ensure proper cardiac function and performance. Metabolism defines a cell context-dependent signature that plays a critical role in survival, proliferation, or differentiation, being a recognized master piece of organ biology, modulating homeostasis, disease progression, and adaptation to tissue damage. The heart is a highly demanding organ, and adult cardiomyocytes require large amount of energy to fulfill adequate contractility. However, functioning under oxidative mitochondrial metabolism is accompanied with a concomitant elevation of harmful reactive oxygen species that indeed contributes to the progression of several cardiovascular pathologies and hampers the regenerative capacity of the mammalian heart. Cardiac metabolism is dynamic along embryonic development and substantially changes as cardiomyocytes mature and differentiate within the first days after birth. During early stages of cardiogenesis, anaerobic glycolysis is the main energetic program, while a progressive switch toward oxidative phosphorylation is a hallmark of myocardium differentiation. In response to cardiac injury, different signaling pathways participate in a metabolic rewiring to reactivate embryonic bioenergetic programs or the utilization of alternative substrates, reflecting the flexibility of heart metabolism and its central role in organ adaptation to external factors. Despite the well-established metabolic pattern of fetal, neonatal, and adult cardiomyocytes, our knowledge about the bioenergetics of other cardiac populations like endothelial cells, cardiac fibroblasts, or immune cells is limited. Considering the close intercellular communication and the influence of nonautonomous cues during heart development and after cardiac damage, it will be fundamental to better understand the metabolic programs in different cardiac cells in order to develop novel interventional opportunities based on metabolic rewiring to prevent heart failure and improve the limited regenerative capacity of the mammalian heart.
Collapse
Affiliation(s)
- Silvia Martin-Puig
- Department of Metabolic and Immune Diseases, Institute for Biomedical Research "Sols-Morreale", National Spanish Research Council, CSIC, Madrid, Spain.
- Cardiac Regeneration Program, National Center for Cardiovascular Research, CNIC, Madrid, Spain.
| | - Ivan Menendez-Montes
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
42
|
Wei J, Duan X, Chen J, Zhang D, Xu J, Zhuang J, Wang S. Metabolic adaptations in pressure overload hypertrophic heart. Heart Fail Rev 2024; 29:95-111. [PMID: 37768435 DOI: 10.1007/s10741-023-10353-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
This review article offers a detailed examination of metabolic adaptations in pressure overload hypertrophic hearts, a condition that plays a pivotal role in the progression of heart failure with preserved ejection fraction (HFpEF) to heart failure with reduced ejection fraction (HFrEF). The paper delves into the complex interplay between various metabolic pathways, including glucose metabolism, fatty acid metabolism, branched-chain amino acid metabolism, and ketone body metabolism. In-depth insights into the shifts in substrate utilization, the role of different transporter proteins, and the potential impact of hypoxia-induced injuries are discussed. Furthermore, potential therapeutic targets and strategies that could minimize myocardial injury and promote cardiac recovery in the context of pressure overload hypertrophy (POH) are examined. This work aims to contribute to a better understanding of metabolic adaptations in POH, highlighting the need for further research on potential therapeutic applications.
Collapse
Affiliation(s)
- Jinfeng Wei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xuefei Duan
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jiaying Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Dengwen Zhang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jindong Xu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Sheng Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Linzhi People's Hospital, Linzhi, Tibet, China.
| |
Collapse
|
43
|
Cao Y, Zeng T, Han W, Ma X, Gu T, Chen L, Tian Y, Xu W, Yin J, Li G, Lu L, Gun S. Comparative analysis of liver transcriptome reveals adaptive responses to hypoxia environmental condition in Tibetan chicken. Anim Biosci 2024; 37:28-38. [PMID: 37641844 PMCID: PMC10766467 DOI: 10.5713/ab.23.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/25/2023] [Accepted: 07/11/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE Tibetan chickens, which have unique adaptations to extreme high-altitude environments, exhibit phenotypic and physiological characteristics that are distinct from those of lowland chickens. However, the mechanisms underlying hypoxic adaptation in the liver of chickens remain unknown. METHODS RNA-sequencing (RNA-Seq) technology was used to assess the differentially expressed genes (DEGs) involved in hypoxia adaptation in highland chickens (native Tibetan chicken [HT]) and lowland chickens (Langshan chicken [LS], Beijing You chicken [BJ], Qingyuan Partridge chicken [QY], and Chahua chicken [CH]). RESULTS A total of 352 co-DEGs were specifically screened between HT and four native lowland chicken breeds. Gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses indicated that these co-DEGs were widely involved in lipid metabolism processes, such as the peroxisome proliferator-activated receptors (PPAR) signaling pathway, fatty acid degradation, fatty acid metabolism and fatty acid biosynthesis. To further determine the relationship from the 352 co-DEGs, protein-protein interaction network was carried out and identified eight genes (ACSL1, CPT1A, ACOX1, PPARC1A, SCD, ACSBG2, ACACA, and FASN) as the potential regulating genes that are responsible for the altitude difference between the HT and other four lowland chicken breeds. CONCLUSION This study provides novel insights into the molecular mechanisms regulating hypoxia adaptation via lipid metabolism in Tibetan chickens and other highland animals.
Collapse
Affiliation(s)
- Yongqing Cao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070,
China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021,
China
| | - Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021,
China
| | - Wei Han
- Jiangsu Institute of Poultry Science, Yangzhou 225125,
China
- Technology Innovation Co., Ltd., Jiangsu Institute of Poultry Science, Yangzhou 211412,
China
| | - Xueying Ma
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy Agricultural and Animal Husbandry Sciences, Lhasa 850004,
China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021,
China
| | - Li Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021,
China
- China-Ukraine Joint Research Center for Protection, Exploitation and Utilization of Poultry Germplasm Resources, Hangzhou 310021,
China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021,
China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021,
China
| | - Jianmei Yin
- Jiangsu Institute of Poultry Science, Yangzhou 225125,
China
- Technology Innovation Co., Ltd., Jiangsu Institute of Poultry Science, Yangzhou 211412,
China
| | - Guohui Li
- Jiangsu Institute of Poultry Science, Yangzhou 225125,
China
- Technology Innovation Co., Ltd., Jiangsu Institute of Poultry Science, Yangzhou 211412,
China
| | - Lizhi Lu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070,
China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou 310021,
China
- China-Ukraine Joint Research Center for Protection, Exploitation and Utilization of Poultry Germplasm Resources, Hangzhou 310021,
China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070,
China
| |
Collapse
|
44
|
Langbøl M, Rovelt J, Saruhanian A, Saruhanian S, Tiedemann D, Baskaran T, Bocca C, Vohra R, Cvenkel B, Lenaers G, Kolko M. Distinct Metabolic Profiles of Ocular Hypertensives in Response to Hypoxia. Int J Mol Sci 2023; 25:195. [PMID: 38203366 PMCID: PMC10779258 DOI: 10.3390/ijms25010195] [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: 11/26/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Glaucoma is a neurodegenerative disease that affects the retinal ganglion cells (RGCs). The main risk factor is elevated intraocular pressure (IOP), but the actual cause of the disease remains unknown. Emerging evidence indicates that metabolic dysfunction plays a central role. The aim of the current study was to determine and compare the effect of universal hypoxia on the metabolomic signature in plasma samples from healthy controls (n = 10), patients with normal-tension glaucoma (NTG, n = 10), and ocular hypertension (OHT, n = 10). By subjecting humans to universal hypoxia, we aim to mimic a state in which the mitochondria in the body are universally stressed. Participants were exposed to normobaric hypoxia for two hours, followed by a 30 min recovery period in normobaric normoxia. Blood samples were collected at baseline, during hypoxia, and in recovery. Plasma samples were analyzed using a non-targeted metabolomics approach based on liquid chromatography coupled with high-resolution mass spectrometry (LC-HRMS). Multivariate analyses were conducted using principal component analysis (PCA) and orthogonal partial least squares-discriminant analysis (OPLS-DA), and univariate analysis using the Wilcoxon signed-rank test and false discovery rate (FDR) correction. Unique metabolites involved in fatty acid biosynthesis and ketone body metabolism were upregulated, while metabolites of the kynurenine pathway were downregulated in OHT patients exposed to universal hypoxia. Differential affection of metabolic pathways may explain why patients with OHT initially do not suffer or are more resilient from optic nerve degeneration. The metabolomes of NTG and OHT patients are regulated differently from control subjects and show dysregulation of metabolites important for energy production. These dysregulated processes may potentially contribute to the elevation of IOP and, ultimately, cell death of the RGCs.
Collapse
Affiliation(s)
- Mia Langbøl
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
| | - Jens Rovelt
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
| | - Arevak Saruhanian
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
| | - Sarkis Saruhanian
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
- Department of Veterinary & Animal Sciences, University of Copenhagen, 2000 Frederiksberg, Denmark
| | - Daniel Tiedemann
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
| | - Thisayini Baskaran
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
| | - Cinzia Bocca
- Faculté de Santé, Institut MITOVASC, UMR CNRS 6015, INSERM U1083, Université d’Angers, 49933 Angers, France; (C.B.); (G.L.)
- Département de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire (CHU), 49933 Angers, France
| | - Rupali Vohra
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
| | - Barbara Cvenkel
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Guy Lenaers
- Faculté de Santé, Institut MITOVASC, UMR CNRS 6015, INSERM U1083, Université d’Angers, 49933 Angers, France; (C.B.); (G.L.)
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (M.L.); (J.R.); (A.S.); (S.S.); (D.T.); (T.B.); (R.V.)
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
| |
Collapse
|
45
|
Kudo T, Zhao ML, Jeknić S, Kovary KM, LaGory EL, Covert MW, Teruel MN. Context-dependent regulation of lipid accumulation in adipocytes by a HIF1α-PPARγ feedback network. Cell Syst 2023; 14:1074-1086.e7. [PMID: 37995680 PMCID: PMC11251692 DOI: 10.1016/j.cels.2023.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 12/03/2022] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
Hypoxia-induced upregulation of HIF1α triggers adipose tissue dysfunction and insulin resistance in obese patients. HIF1α closely interacts with PPARγ, the master regulator of adipocyte differentiation and lipid accumulation, but there are conflicting results regarding how this interaction controls the excessive lipid accumulation that drives adipocyte dysfunction. To directly address these conflicts, we established a differentiation system that recapitulated prior seemingly opposing observations made across different experimental settings. Using single-cell imaging and coarse-grained mathematical modeling, we show how HIF1α can both promote and repress lipid accumulation during adipogenesis. Our model predicted and our experiments confirmed that the opposing roles of HIF1α are isolated from each other by the positive-feedback-mediated upregulation of PPARγ that drives adipocyte differentiation. Finally, we identify three factors: strength of the differentiation cue, timing of hypoxic perturbation, and strength of HIF1α expression changes that, when considered together, provide an explanation for many of the previous conflicting reports.
Collapse
Affiliation(s)
- Takamasa Kudo
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Michael L Zhao
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Stevan Jeknić
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kyle M Kovary
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Edward L LaGory
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Markus W Covert
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | - Mary N Teruel
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Biochemistry and the Drukier Institute of Children's Health, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
46
|
Di Florio D, Gorelov D, McCabe E, Beetler D, Shapiro K, Bruno K, Chekuri I, Jain A, Whelan E, Salomon G, Khatib S, Bonvie-Hill N, Giresi P, Balamurugan V, Weigel G, Fliess J, Darakjian A, Edenfield B, Kocsis C, McLeod C, Cooper L, Audet-Walsh E, Coronado M, Sin J, Fairweather D. Sex differences in mitochondrial gene expression during viral myocarditis. RESEARCH SQUARE 2023:rs.3.rs-3716881. [PMID: 38196574 PMCID: PMC10775395 DOI: 10.21203/rs.3.rs-3716881/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Background Myocarditis is an inflammation of the heart muscle most often caused by an immune response to viral infections. Sex differences in the immune response during myocarditis have been well described but upstream mechanisms in the heart that might influence sex differences in disease are not completely understood. Methods Male and female BALB/c wild type mice received an intraperitoneal injection of heart-passaged coxsackievirus B3 (CVB3) or vehicle control. Bulk-tissue RNA-sequencing was conducted to better understand sex differences in CVB3 myocarditis. We performed enrichment analysis to understand sex differences in the transcriptional landscape of myocarditis and identify candidate transcription factors that might drive sex differences in myocarditis. Results The hearts of male and female mice with myocarditis were significantly enriched for pathways related to an innate and adaptive immune response compared to uninfected controls. When comparing females to males with myocarditis, males were enriched for inflammatory pathways and gene changes that suggested worse mitochondrial transcriptional support (e.g., mitochondrial electron transport genes). In contrast, females were enriched for pathways related to mitochondrial respiration and bioenergetics, which were confirmed by higher transcript levels of master regulators of mitochondrial function including peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1α), nuclear respiratory factor 1 (NRF1) and estrogen-related receptor alpha (ERRα). TRANSFAC analysis identified ERRa as a transcription factor that may mediate sex differences in mitochondrial function during myocarditis. Conclusions Master regulators of mitochondrial function were elevated in females with myocarditis compared to males and may promote sex differences in mitochondrial respiratory transcript expression during viral myocarditis resulting in less severe myocarditis in females following viral infection.
Collapse
|
47
|
Szibor M, Mühlon M, Doenst T, Pohjoismäki JLO. Spatial adjustment of bioenergetics, a possible determinant of contractile adaptation and development of contractile failure. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1305960. [PMID: 39086691 PMCID: PMC11285667 DOI: 10.3389/fmmed.2023.1305960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/23/2023] [Indexed: 08/02/2024]
Abstract
Cardiomyocytes depend on mitochondrial oxidative phosphorylation (OXPHOS) for energy metabolism, which is facilitated by the mitochondrial electron transfer system (ETS). In a series of thermogenic redox reactions, electrons are shuttled through the ETS to oxygen as the final electron acceptor. This electron transfer is coupled to proton translocation across the inner mitochondrial membrane, which itself is the main driving force for ATP production. Oxygen availability is thus a prerequisite for ATP production and consequently contractility. Notably, cardiomyocytes are exceptionally large cells and densely packed with contractile structures, which constrains intracellular oxygen distribution. Moreover, oxygen must pass through layers of actively respiring mitochondria to reach the ones located in the innermost contractile compartment. Indeed, uneven oxygen distribution was observed in cardiomyocytes, suggesting that local ATP supply may also vary according to oxygen availability. Here, we discuss how spatial adjustment of bioenergetics to intracellular oxygen fluctuations may underlie cardiac contractile adaptation and how this adaptation may pose a risk for the development of contractile failure.
Collapse
Affiliation(s)
- Marten Szibor
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich-Schiller University of Jena, Jena, Germany
- BioMediTech and Tampere University Hospital, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marie Mühlon
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich-Schiller University of Jena, Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich-Schiller University of Jena, Jena, Germany
| | - Jaakko L. O. Pohjoismäki
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
48
|
Owesny P, Grune T. The link between obesity and aging - insights into cardiac energy metabolism. Mech Ageing Dev 2023; 216:111870. [PMID: 37689316 DOI: 10.1016/j.mad.2023.111870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Obesity and aging are well-established risk factors for a range of diseases, including cardiovascular diseases and type 2 diabetes. Given the escalating prevalence of obesity, the aging population, and the subsequent increase in cardiovascular diseases, it is crucial to investigate the underlying mechanisms involved. Both aging and obesity have profound effects on the energy metabolism through various mechanisms, including metabolic inflexibility, altered substrate utilization for energy production, deregulated nutrient sensing, and mitochondrial dysfunction. In this review, we aim to present and discuss the hypothesis that obesity, due to its similarity in changes observed in the aging heart, may accelerate the process of cardiac aging and exacerbate the clinical outcomes of elderly individuals with obesity.
Collapse
Affiliation(s)
- Patricia Owesny
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| |
Collapse
|
49
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
50
|
Corradi F, Masini G, Bucciarelli T, De Caterina R. Iron deficiency in myocardial ischaemia: molecular mechanisms and therapeutic perspectives. Cardiovasc Res 2023; 119:2405-2420. [PMID: 37722377 DOI: 10.1093/cvr/cvad146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/14/2023] [Accepted: 07/10/2023] [Indexed: 09/20/2023] Open
Abstract
Systemic iron deficiency (SID), even in the absence of anaemia, worsens the prognosis and increases mortality in heart failure (HF). Recent clinical-epidemiological studies, however, have shown that a myocardial iron deficiency (MID) is frequently present in cases of severe HF, even in the absence of SID and without anaemia. In addition, experimental studies have shown a poor correlation between the state of systemic and myocardial iron. MID in animal models leads to severe mitochondrial dysfunction, alterations of mitophagy, and mitochondrial biogenesis, with profound alterations in cardiac mechanics and the occurrence of a fatal cardiomyopathy, all effects prevented by intravenous administration of iron. This shifts the focus to the myocardial state of iron, in the absence of anaemia, as an important factor in prognostic worsening and mortality in HF. There is now epidemiological evidence that SID worsens prognosis and mortality also in patients with acute and chronic coronary heart disease and experimental evidence that MID aggravates acute myocardial ischaemia as well as post-ischaemic remodelling. Intravenous administration of ferric carboxymaltose (FCM) or ferric dextrane improves post-ischaemic adverse remodelling. We here review such evidence, propose that MID worsens ischaemia/reperfusion injury, and discuss possible molecular mechanisms, such as chronic hyperactivation of HIF1-α, exacerbation of cytosolic and mitochondrial calcium overload, amplified increase of mitochondrial [NADH]/[NAD+] ratio, and depletion of energy status and NAD+ content with inhibition of sirtuin 1-3 activity. Such evidence now portrays iron metabolism as a core factor not only in HF but also in myocardial ischaemia.
Collapse
Affiliation(s)
- Francesco Corradi
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, Via dei Vestini, 66100, Chieti, Italy
| | - Gabriele Masini
- Chair and Postgraduate School of Cardiology, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Tonino Bucciarelli
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, Via dei Vestini, 66100, Chieti, Italy
| | - Raffaele De Caterina
- Chair and Postgraduate School of Cardiology, University of Pisa, Via Savi 10, 56126, Pisa, Italy
- Fondazione VillaSerena per la Ricerca, Viale L. Petruzzi 42, 65013, Città Sant'Angelo, Pescara, Italy
| |
Collapse
|