1
|
Moka MK, K SD, George M. "Emerging clinical approaches in diabetic cardiomyopathy: insights from clinical trials and future directions". Acta Diabetol 2024:10.1007/s00592-024-02363-5. [PMID: 39254745 DOI: 10.1007/s00592-024-02363-5] [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: 06/19/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
AIM We aim to explore the potential of diverse treatments, including perhexiline, calcium channel blockers, anti-hypertensives, PDE5 inhibitors, anti-anginal drugs, aldose reductase inhibitors, and SGLT-2 inhibitors, supported by clinical evidence. Additionally, this review seeks to identify novel therapeutic targets and future avenues for improving cardiovascular outcomes in diabetic populations. METHOD We performed a comprehensive literature review of English-language studies across multiple electronic databases, such as PubMed, ScienceDirect, Scopus, and Google Scholar, focusing on clinical trials. The search utilized keywords including 'Anti-hyperglycaemic drug,' 'Diabetic cardiomyopathy,' 'DPP-4 inhibitors,' 'GLP-1 receptor agonists,' 'Heart failure,' and 'SGLT-2 inhibitors.' RESULT We assessed clinical investigations in the treatment of cardiomyopathy and diabetes mellitus (DM) that are enhancing our understanding through trials evaluating the Polypill, Perhexiline, Eplerenone, IMB-1018972, AT-001, tadalafil, and dapagliflozin inhibitors. The development of new targeted interventions is of paramount importance due to the overlooked early symptoms, the complexity of the cellular and molecular pathways involved, and the absence of effective drug therapies. CONCLUSION Pharmacological treatments like GLP-1 agonists, SGLT-2 inhibitors, NHE-1, NHE-3, and PPAR-γ agonists show promise for treating DCM. These treatments improve myocardial glucose absorption, address dysregulated glucose and lipid metabolism, and lower heart failure and cardiovascular events. Further research is needed to confirm effectiveness and safety.
Collapse
Affiliation(s)
- Murali Krishna Moka
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India
- Department of Diabetology and Endocrinology, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India
| | - Sriram D K
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India
- Department of Diabetology and Endocrinology, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India
| | - Melvin George
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India.
- Department of Diabetology and Endocrinology, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India.
| |
Collapse
|
2
|
Heather LC, Gopal K, Srnic N, Ussher JR. Redefining Diabetic Cardiomyopathy: Perturbations in Substrate Metabolism at the Heart of Its Pathology. Diabetes 2024; 73:659-670. [PMID: 38387045 PMCID: PMC11043056 DOI: 10.2337/dbi23-0019] [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: 07/27/2023] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Cardiovascular disease represents the leading cause of death in people with diabetes, most notably from macrovascular diseases such as myocardial infarction or heart failure. Diabetes also increases the risk of a specific form of cardiomyopathy, referred to as diabetic cardiomyopathy (DbCM), originally defined as ventricular dysfunction in the absence of underlying coronary artery disease and/or hypertension. Herein, we provide an overview on the key mediators of DbCM, with an emphasis on the role for perturbations in cardiac substrate metabolism. We discuss key mechanisms regulating metabolic dysfunction in DbCM, with additional focus on the role of metabolites as signaling molecules within the diabetic heart. Furthermore, we discuss the preclinical approaches to target these perturbations to alleviate DbCM. With several advancements in our understanding, we propose the following as a new definition for, or approach to classify, DbCM: "diastolic dysfunction in the presence of altered myocardial metabolism in a person with diabetes but absence of other known causes of cardiomyopathy and/or hypertension." However, we recognize that no definition can fully explain the complexity of why some individuals with DbCM exhibit diastolic dysfunction, whereas others develop systolic dysfunction. Due to DbCM sharing pathological features with heart failure with preserved ejection fraction (HFpEF), the latter of which is more prevalent in the population with diabetes, it is imperative to determine whether effective management of DbCM decreases HFpEF prevalence. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Lisa C. Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Nikola Srnic
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - John R. Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Lin QR, Jia LQ, Lei M, Gao D, Zhang N, Sha L, Liu XH, Liu YD. Natural products as pharmacological modulators of mitochondrial dysfunctions for the treatment of diabetes and its complications: An update since 2010. Pharmacol Res 2024; 200:107054. [PMID: 38181858 DOI: 10.1016/j.phrs.2023.107054] [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: 09/25/2023] [Revised: 12/12/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024]
Abstract
Diabetes, characterized as a well-known chronic metabolic syndrome, with its associated complications pose a substantial and escalating health and healthcare challenge on a global scale. Current strategies addressing diabetes are mainly symptomatic and there are fewer available curative pharmaceuticals for diabetic complications. Thus, there is an urgent need to identify novel pharmacological targets and agents. The impaired mitochondria have been associated with the etiology of diabetes and its complications, and the intervention of mitochondrial dysfunction represents an attractive breakthrough point for the treatments of diabetes and its complications. Natural products (NPs), with multicenter characteristics, multi-pharmacological activities and lower toxicity, have been caught attentions as the modulators of mitochondrial functions in the therapeutical filed of diabetes and its complications. This review mainly summarizes the recent progresses on the potential of 39 NPs and 2 plant-extracted mixtures to improve mitochondrial dysfunction against diabetes and its complications. It is expected that this work may be useful to accelerate the development of innovative drugs originated from NPs and improve upcoming therapeutics in diabetes and its complications.
Collapse
Affiliation(s)
- Qian-Ru Lin
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Lian-Qun Jia
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 116600, China
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Di Gao
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Nan Zhang
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Lei Sha
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Xu-Han Liu
- Department of Endocrinology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, China.
| | - Yu-Dan Liu
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
4
|
Hu M, Jiang W, Ye C, Hu T, Yu Q, Meng M, Sun L, Liang J, Chen Y. Honokiol attenuates high glucose-induced peripheral neuropathy via inhibiting ferroptosis and activating AMPK/SIRT1/PGC-1α pathway in Schwann cells. Phytother Res 2023; 37:5787-5802. [PMID: 37580045 DOI: 10.1002/ptr.7984] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/16/2023]
Abstract
Schwann cells injury induced by high glucose (HG) contributes to the development of diabetic peripheral neuropathy (DPN). Honokiol has been reported to regulate glucose metabolism, however, its effect on DPN and the precise molecular mechanisms remain unclear. This study aimed to investigate the role of AMPK/SIRT1/PGC-1α axis in the protective effects of honokiol on DPN. The biochemical assay and JC-1 staining results demonstrated that honokiol reduced HG-induced oxidative stress and ferroptosis as well as mitochondrial dysfunction in Schwann cells. RT-qPCR and western blotting were utilized to investigate the mechanism of action of honokiol, and the results showed that HG-induced inhibition of AMPK/SIRT1/PGC-1α axis and changes of downstream gene expression profile were restored by honokiol. Moreover, silencing of Sirt1 by siRNA delivery markedly diminished the changes of gene expression profile induced by honokiol in HG-induced Schwann cells. More importantly, we found that administration of honokiol remarkably attenuated DPN via improving sciatic nerve conduction velocity and increasing thermal and mechanical sensitivity in streptozotocin-induced diabetic rats. Collectively, these results demonstrate that honokiol can attenuate HG-induced Schwann cells injury and peripheral nerve dysfunction, suggesting a novel potential strategy for treatment of DPN.
Collapse
Affiliation(s)
- Man Hu
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Wen Jiang
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Chen Ye
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Ting Hu
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Qingqing Yu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Moran Meng
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Lijuan Sun
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Jichao Liang
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Yong Chen
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
5
|
Khatoon F, Ali S, Kumar V, Elasbali AM, Alhassan HH, Alharethi SH, Islam A, Hassan MI. Pharmacological features, health benefits and clinical implications of honokiol. J Biomol Struct Dyn 2023; 41:7511-7533. [PMID: 36093963 DOI: 10.1080/07391102.2022.2120541] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
Abstract
Honokiol (HNK) is a natural polyphenolic compound extracted from the bark and leaves of Magnolia grandiflora. It has been traditionally used as a medicinal compound to treat inflammatory diseases. HNK possesses numerous health benefits with a minimal level of toxicity. It can cross the blood-brain barrier and blood-cerebrospinal fluid, thus having significant bioavailability in the neurological tissues. HNK is a promising bioactive compound possesses neuroprotective, antimicrobial, anti-tumorigenic, anti-spasmodic, antidepressant, analgesic, and antithrombotic features . HNK can prevent the growth of several cancer types and haematological malignancies. Recent studies suggested its role in COVID-19 therapy. It binds effectively with several molecular targets, including apoptotic factors, chemokines, transcription factors, cell surface adhesion molecules, and kinases. HNK has excellent pharmacological features and a wide range of chemotherapeutic effects, and thus, researchers have increased interest in improving the therapeutic implications of HNK to the clinic as a novel agent. This review focused on the therapeutic implications of HNK, highlighting clinical and pharmacological features and the underlying mechanism of action.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatima Khatoon
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Hassan H Alhassan
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran, Saudia Arabia
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
6
|
Gupta A. Cardiac 31P MR spectroscopy: development of the past five decades and future vision-will it be of diagnostic use in clinics? Heart Fail Rev 2023; 28:485-532. [PMID: 36427161 DOI: 10.1007/s10741-022-10287-x] [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] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
In the past five decades, the use of the magnetic resonance (MR) technique for cardiovascular diseases has engendered much attention and raised the opportunity that the technique could be useful for clinical applications. MR has two arrows in its quiver: One is magnetic resonance imaging (MRI), and the other is magnetic resonance spectroscopy (MRS). Non-invasively, highly advanced MRI provides unique and profound information about the anatomical changes of the heart. Excellently developed MRS provides irreplaceable and insightful evidence of the real-time biochemistry of cardiac metabolism of underpinning diseases. Compared to MRI, which has already been successfully applied in routine clinical practice, MRS still has a long way to travel to be incorporated into routine diagnostics. Considering the exceptional potential of 31P MRS to measure the real-time metabolic changes of energetic molecules qualitatively and quantitatively, how far its powerful technique should be waited before a successful transition from "bench-to-bedside" is enticing. The present review highlights the seminal studies on the chronological development of cardiac 31P MRS in the past five decades and the future vision and challenges to incorporating it for routine diagnostics of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Gupta
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow, 226014, India.
| |
Collapse
|
7
|
Capone F, Sotomayor-Flores C, Bode D, Wang R, Rodolico D, Strocchi S, Schiattarella GG. Cardiac metabolism in HFpEF: from fuel to signalling. Cardiovasc Res 2023; 118:3556-3575. [PMID: 36504368 DOI: 10.1093/cvr/cvac166] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 12/14/2022] Open
Abstract
Heart failure (HF) is marked by distinctive changes in myocardial uptake and utilization of energy substrates. Among the different types of HF, HF with preserved ejection fraction (HFpEF) is a highly prevalent, complex, and heterogeneous condition for which metabolic derangements seem to dictate disease progression. Changes in intermediate metabolism in cardiometabolic HFpEF-among the most prevalent forms of HFpEF-have a large impact both on energy provision and on a number of signalling pathways in the heart. This dual, metabolic vs. signalling, role is played in particular by long-chain fatty acids (LCFAs) and short-chain carbon sources [namely, short-chain fatty acids (SCFAs) and ketone bodies (KBs)]. LCFAs are key fuels for the heart, but their excess can be harmful, as in the case of toxic accumulation of lipid by-products (i.e. lipotoxicity). SCFAs and KBs have been proposed as a potential major, alternative source of energy in HFpEF. At the same time, both LCFAs and short-chain carbon sources are substrate for protein post-translational modifications and other forms of direct and indirect signalling of pivotal importance in HFpEF pathogenesis. An in-depth molecular understanding of the biological functions of energy substrates and their signalling role will be instrumental in the development of novel therapeutic approaches to HFpEF. Here, we summarize the current evidence on changes in energy metabolism in HFpEF, discuss the signalling role of intermediate metabolites through, at least in part, their fate as substrates for post-translational modifications, and highlight clinical and translational challenges around metabolic therapy in HFpEF.
Collapse
Affiliation(s)
- Federico Capone
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Division of Internal Medicine, Department of Medicine, University of Padua, Padua, Italy
| | - Cristian Sotomayor-Flores
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - David Bode
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Rongling Wang
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Daniele Rodolico
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Stefano Strocchi
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
8
|
Liu H, Luo W, Liu J, Kang X, Yan J, Zhang T, Yang L, Shen L, Liu D. The glucotoxicity protecting effect of honokiol in human hepatocytes via directly activating AMPK. Front Nutr 2022; 9:1043009. [DOI: 10.3389/fnut.2022.1043009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
IntroductionSustained hyperglycemia causes glucotoxicity, which has been regarded as a contributor to hepatocyte damage in type 2 diabetes (T2D) and its metabolic comorbidities. Honokiol is a natural biphenolic component derived from the dietary supplement Magnolia officinalis extract. This study aimed to investigate the effects of honokiol on glucose metabolism disorders and oxidative stress in hepatocytes and the underlying mechanisms.MethodsHepG2 cells were treated with glucosamines (18 mM) to induce glucotoxicity as a diabetic complication model in vitro.Results and discussionHonokiol significantly increased glucose consumption, elevated 2-NBDG uptake, and promoted GLUT2 translocation to the plasma membrane in glucosamine-treated HepG2 cells, indicating that honokiol ameliorates glucose metabolism disorders. Furthermore, glucosamine-induced ROS accumulation and loss of mitochondrial membrane potential were markedly reduced by honokiol, suggesting that honokiol alleviated glucotoxicity-induced oxidative stress. These effects were largely abolished by compound C, an AMPK inhibitor, suggesting an AMPK activation-dependent manner of honokiol function in promoting glucose metabolism and mitigating oxidative stress. Molecular docking results revealed that honokiol could interact with the amino acid residues (His151, Arg152, Lys243, Arg70, Lys170, and His298) in the active site of AMPK. These findings provide new insights into the antidiabetic effect of honokiol, which may be a promising agent for the prevention and treatment of T2D and associated metabolic comorbidities.
Collapse
|
9
|
Park S, Mossmann D, Chen Q, Wang X, Dazert E, Colombi M, Schmidt A, Ryback B, Ng CKY, Terracciano LM, Heim MH, Hall MN. Transcription factors TEAD2 and E2A globally repress acetyl-CoA synthesis to promote tumorigenesis. Mol Cell 2022; 82:4246-4261.e11. [PMID: 36400009 DOI: 10.1016/j.molcel.2022.10.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/22/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022]
Abstract
Acetyl-coenzyme A (acetyl-CoA) plays an important role in metabolism, gene expression, signaling, and other cellular processes via transfer of its acetyl group to proteins and metabolites. However, the synthesis and usage of acetyl-CoA in disease states such as cancer are poorly characterized. Here, we investigated global acetyl-CoA synthesis and protein acetylation in a mouse model and patient samples of hepatocellular carcinoma (HCC). Unexpectedly, we found that acetyl-CoA levels are decreased in HCC due to transcriptional downregulation of all six acetyl-CoA biosynthesis pathways. This led to hypo-acetylation specifically of non-histone proteins, including many enzymes in metabolic pathways. Importantly, repression of acetyl-CoA synthesis promoted oncogenic dedifferentiation and proliferation. Mechanistically, acetyl-CoA synthesis was repressed by the transcription factors TEAD2 and E2A, previously unknown to control acetyl-CoA synthesis. Knockdown of TEAD2 and E2A restored acetyl-CoA levels and inhibited tumor growth. Our findings causally link transcriptional reprogramming of acetyl-CoA metabolism, dedifferentiation, and cancer.
Collapse
Affiliation(s)
- Sujin Park
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Dirk Mossmann
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Qian Chen
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland; Division of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, 4031 Basel, Switzerland
| | - Xueya Wang
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland; Division of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, 4031 Basel, Switzerland
| | - Eva Dazert
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Marco Colombi
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | | | - Brendan Ryback
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Charlotte K Y Ng
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Markus H Heim
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland; Division of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, 4031 Basel, Switzerland
| | - Michael N Hall
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
10
|
Pérez-Hernández M, van Opbergen CJM, Bagwan N, Vissing CR, Marrón-Liñares GM, Zhang M, Torres Vega E, Sorrentino A, Drici L, Sulek K, Zhai R, Hansen FB, Christensen AH, Boesgaard S, Gustafsson F, Rossing K, Small EM, Davies MJ, Rothenberg E, Sato PY, Cerrone M, Jensen THL, Qvortrup K, Bundgaard H, Delmar M, Lundby A. Loss of Nuclear Envelope Integrity and Increased Oxidant Production Cause DNA Damage in Adult Hearts Deficient in PKP2: A Molecular Substrate of ARVC. Circulation 2022; 146:851-867. [PMID: 35959657 PMCID: PMC9474627 DOI: 10.1161/circulationaha.122.060454] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/30/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Arrhythmogenic right ventricular cardiomyopathy (ARVC) is characterized by high propensity to life-threatening arrhythmias and progressive loss of heart muscle. More than 40% of reported genetic variants linked to ARVC reside in the PKP2 gene, which encodes the PKP2 protein (plakophilin-2). METHODS We describe a comprehensive characterization of the ARVC molecular landscape as determined by high-resolution mass spectrometry, RNA sequencing, and transmission electron microscopy of right ventricular biopsy samples obtained from patients with ARVC with PKP2 mutations and left ventricular ejection fraction >45%. Samples from healthy relatives served as controls. The observations led to experimental work using multiple imaging and biochemical techniques in mice with a cardiac-specific deletion of Pkp2 studied at a time of preserved left ventricular ejection fraction and in human induced pluripotent stem cell-derived PKP2-deficient myocytes. RESULTS Samples from patients with ARVC present a loss of nuclear envelope integrity, molecular signatures indicative of increased DNA damage, and a deficit in transcripts coding for proteins in the electron transport chain. Mice with a cardiac-specific deletion of Pkp2 also present a loss of nuclear envelope integrity, which leads to DNA damage and subsequent excess oxidant production (O2.- and H2O2), the latter increased further under mechanical stress (isoproterenol or exercise). Increased oxidant production and DNA damage is recapitulated in human induced pluripotent stem cell-derived PKP2-deficient myocytes. Furthermore, PKP2-deficient cells release H2O2 into the extracellular environment, causing DNA damage and increased oxidant production in neighboring myocytes in a paracrine manner. Treatment with honokiol increases SIRT3 (mitochondrial nicotinamide adenine dinucleotide-dependent protein deacetylase sirtuin-3) activity, reduces oxidant levels and DNA damage in vitro and in vivo, reduces collagen abundance in the right ventricular free wall, and has a protective effect on right ventricular function. CONCLUSIONS Loss of nuclear envelope integrity and subsequent DNA damage is a key substrate in the molecular pathology of ARVC. We show transcriptional downregulation of proteins of the electron transcript chain as an early event in the molecular pathophysiology of the disease (before loss of left ventricular ejection fraction <45%), which associates with increased oxidant production (O2.- and H2O2). We propose therapies that limit oxidant formation as a possible intervention to restrict DNA damage in ARVC.
Collapse
Affiliation(s)
- Marta Pérez-Hernández
- The Leon H. Charney Division of Cardiology, NYU-Grossman School of Medicine, New York (M.P.-H., C.J.M.v.O., G.M.M.-L., M.Z., M.C., M.D.)
| | - Chantal J M van Opbergen
- The Leon H. Charney Division of Cardiology, NYU-Grossman School of Medicine, New York (M.P.-H., C.J.M.v.O., G.M.M.-L., M.Z., M.C., M.D.)
| | - Navratan Bagwan
- Department of Biomedical Sciences (N.B., E.T.V., A.S., F.B.H., M.J.D., K.Q., A.L.), University of Copenhagen, Denmark
| | - Christoffer Rasmus Vissing
- Faculty of Health and Medical Sciences, and Department of Clinical Medicine (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.), University of Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.)
| | - Grecia M Marrón-Liñares
- The Leon H. Charney Division of Cardiology, NYU-Grossman School of Medicine, New York (M.P.-H., C.J.M.v.O., G.M.M.-L., M.Z., M.C., M.D.)
| | - Mingliang Zhang
- The Leon H. Charney Division of Cardiology, NYU-Grossman School of Medicine, New York (M.P.-H., C.J.M.v.O., G.M.M.-L., M.Z., M.C., M.D.)
| | - Estefania Torres Vega
- Department of Biomedical Sciences (N.B., E.T.V., A.S., F.B.H., M.J.D., K.Q., A.L.), University of Copenhagen, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences (N.B., E.T.V., A.S., F.B.H., M.J.D., K.Q., A.L.), University of Copenhagen, Denmark
| | - Lylia Drici
- The Novo Nordisk Foundation Center for Protein Research (L.D., K.S.), University of Copenhagen, Denmark
| | - Karolina Sulek
- The Novo Nordisk Foundation Center for Protein Research (L.D., K.S.), University of Copenhagen, Denmark
| | - Ruxu Zhai
- College of Medicine, Drexel University, Philadelphia, PA (R.Z., P.Y.S.)
| | - Finn B Hansen
- Department of Biomedical Sciences (N.B., E.T.V., A.S., F.B.H., M.J.D., K.Q., A.L.), University of Copenhagen, Denmark
| | - Alex H Christensen
- Faculty of Health and Medical Sciences, and Department of Clinical Medicine (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.), University of Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.)
- Department of Cardiology, Copenhagen University Hospital-Herlev-Gentofte Hospital, Denmark (A.H.C.)
| | - Søren Boesgaard
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.)
- College of Medicine, Drexel University, Philadelphia, PA (R.Z., P.Y.S.)
| | - Finn Gustafsson
- Faculty of Health and Medical Sciences, and Department of Clinical Medicine (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.), University of Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.)
| | - Kasper Rossing
- Faculty of Health and Medical Sciences, and Department of Clinical Medicine (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.), University of Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.)
| | - Eric M Small
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, NY (E.M.S.)
| | - Michael J Davies
- Department of Biomedical Sciences (N.B., E.T.V., A.S., F.B.H., M.J.D., K.Q., A.L.), University of Copenhagen, Denmark
| | - Eli Rothenberg
- Division of Pharmacology, NYU School of Medicine, New York (E.R.)
| | - Priscila Y Sato
- College of Medicine, Drexel University, Philadelphia, PA (R.Z., P.Y.S.)
| | - Marina Cerrone
- The Leon H. Charney Division of Cardiology, NYU-Grossman School of Medicine, New York (M.P.-H., C.J.M.v.O., G.M.M.-L., M.Z., M.C., M.D.)
| | - Thomas Hartvig Lindkær Jensen
- Faculty of Health and Medical Sciences, and Department of Clinical Medicine (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.), University of Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.)
| | - Klaus Qvortrup
- Department of Biomedical Sciences (N.B., E.T.V., A.S., F.B.H., M.J.D., K.Q., A.L.), University of Copenhagen, Denmark
| | - Henning Bundgaard
- Faculty of Health and Medical Sciences, and Department of Clinical Medicine (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.), University of Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark (C.R.V., A.H.C., S.B., F.G., K.R., T.H.L.J., H.B.)
| | - Mario Delmar
- The Leon H. Charney Division of Cardiology, NYU-Grossman School of Medicine, New York (M.P.-H., C.J.M.v.O., G.M.M.-L., M.Z., M.C., M.D.)
| | - Alicia Lundby
- Department of Biomedical Sciences (N.B., E.T.V., A.S., F.B.H., M.J.D., K.Q., A.L.), University of Copenhagen, Denmark
| |
Collapse
|
11
|
Heather LC, Hafstad AD, Halade GV, Harmancey R, Mellor KM, Mishra PK, Mulvihill EE, Nabben M, Nakamura M, Rider OJ, Ruiz M, Wende AR, Ussher JR. Guidelines on Models of Diabetic Heart Disease. Am J Physiol Heart Circ Physiol 2022; 323:H176-H200. [PMID: 35657616 PMCID: PMC9273269 DOI: 10.1152/ajpheart.00058.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Diabetes is a major risk factor for cardiovascular diseases, including diabetic cardiomyopathy, atherosclerosis, myocardial infarction, and heart failure. As cardiovascular disease represents the number one cause of death in people with diabetes, there has been a major emphasis on understanding the mechanisms by which diabetes promotes cardiovascular disease, and how antidiabetic therapies impact diabetic heart disease. With a wide array of models to study diabetes (both type 1 and type 2), the field has made major progress in answering these questions. However, each model has its own inherent limitations. Therefore, the purpose of this guidelines document is to provide the field with information on which aspects of cardiovascular disease in the human diabetic population are most accurately reproduced by the available models. This review aims to emphasize the advantages and disadvantages of each model, and to highlight the practical challenges and technical considerations involved. We will review the preclinical animal models of diabetes (based on their method of induction), appraise models of diabetes-related atherosclerosis and heart failure, and discuss in vitro models of diabetic heart disease. These guidelines will allow researchers to select the appropriate model of diabetic heart disease, depending on the specific research question being addressed.
Collapse
Affiliation(s)
- Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anne D Hafstad
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ganesh V Halade
- Department of Medicine, The University of Alabama at Birmingham, Tampa, Florida, United States
| | - Romain Harmancey
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Erin E Mulvihill
- University of Ottawa Heart Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Miranda Nabben
- Departments of Genetics and Cell Biology, and Clinical Genetics, Maastricht University Medical Center, CARIM School of Cardiovascular Diseases, Maastricht, the Netherlands
| | - Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Oliver J Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matthieu Ruiz
- Montreal Heart Institute, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
12
|
Luo Q, Li X, Zhong W, Cao W, Zhu M, Wu A, Chen W, Ye Z, Han Q, Natarajan D, Pathak JL, Zhang Q. Dicalcium silicate-induced mitochondrial dysfunction and autophagy-mediated macrophagic inflammation promotes osteogenic differentiation of BMSCs. Regen Biomater 2021; 9:rbab075. [PMID: 35480858 PMCID: PMC9039510 DOI: 10.1093/rb/rbab075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 11/14/2022] Open
Abstract
Dicalcium silicate (Ca2SiO4, C2S) has osteogenic potential but induces macrophagic inflammation. Mitochondrial function plays a vital role in macrophage polarization and macrophagic inflammation. The mitochondrial function of C2S-treated macrophages is still unclear. This study hypothesized: (i) the C2S modulates mitochondrial function and autophagy in macrophages to regulate macrophagic inflammation, and (ii) C2S-induced macrophagic inflammation regulates osteogenesis. We used RAW264.7 cells as a model of macrophage. The C2S (75-150 μg/ml) extract was used to analyze the macrophagic mitochondrial function and macrophage-mediated effect on osteogenic differentiation of mouse bone marrow-derived mesenchymal stem cells (BMSCs). The results showed that C2S extract (150 μg/ml) induced TNF-α, IL-1β and IL-6 production in macrophages. C2S extract (150 μg/ml) enhanced reactive oxygen species level and intracellular calcium level but reduced mitochondrial membrane potential and ATP production. TEM images showed reduced mitochondrial abundance and altered the mitochondrial morphology in C2S (150 μg/ml)-treated macrophages. Protein level expression of PINK1, Parkin, Beclin1 and LC3 was upregulated but TOMM20 was downregulated. mRNA sequencing and KEGG analysis showed that C2S-induced differentially expressed mRNAs in macrophages were mainly distributed in the essential signaling pathways involved in mitochondrial function and autophagy. The conditioned medium from C2S-treated macrophage robustly promoted osteogenic differentiation in BMSCs. In conclusion, our results indicate mitochondrial dysfunction and autophagy as the possible mechanism of C2S-induced macrophagic inflammation. The promotion of osteogenic differentiation of BMSCs by the C2S-induced macrophagic inflammation suggests the potential application of C2S in developing immunomodulatory bone grafts.
Collapse
Affiliation(s)
- Qianting Luo
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen 529030, China
| | - Xingyang Li
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wenchao Zhong
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wei Cao
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
- Department of Oral Cell Biology, Academic Centre of Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam 1081LA, The Netherlands
| | - Mingjing Zhu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Antong Wu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Wanyi Chen
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Zhitong Ye
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Qiao Han
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Duraipandy Natarajan
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Janak L Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| | - Qingbin Zhang
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou 510182, China
| |
Collapse
|
13
|
Ketema EB, Lopaschuk GD. Post-translational Acetylation Control of Cardiac Energy Metabolism. Front Cardiovasc Med 2021; 8:723996. [PMID: 34409084 PMCID: PMC8365027 DOI: 10.3389/fcvm.2021.723996] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 06/30/2021] [Indexed: 12/17/2022] Open
Abstract
Perturbations in myocardial energy substrate metabolism are key contributors to the pathogenesis of heart diseases. However, the underlying causes of these metabolic alterations remain poorly understood. Recently, post-translational acetylation-mediated modification of metabolic enzymes has emerged as one of the important regulatory mechanisms for these metabolic changes. Nevertheless, despite the growing reports of a large number of acetylated cardiac mitochondrial proteins involved in energy metabolism, the functional consequences of these acetylation changes and how they correlate to metabolic alterations and myocardial dysfunction are not clearly defined. This review summarizes the evidence for a role of cardiac mitochondrial protein acetylation in altering the function of major metabolic enzymes and myocardial energy metabolism in various cardiovascular disease conditions.
Collapse
Affiliation(s)
- Ezra B Ketema
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Gary D Lopaschuk
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Kerr M, Dennis KMJH, Carr CA, Fuller W, Berridge G, Rohling S, Aitken CL, Lopez C, Fischer R, Miller JJ, Clarke K, Tyler DJ, Heather LC. Diabetic mitochondria are resistant to palmitoyl CoA inhibition of respiration, which is detrimental during ischemia. FASEB J 2021; 35:e21765. [PMID: 34318967 PMCID: PMC8662312 DOI: 10.1096/fj.202100394r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/24/2021] [Accepted: 06/14/2021] [Indexed: 01/07/2023]
Abstract
The bioactive lipid intermediate palmitoyl CoA (PCoA) can inhibit mitochondrial ADP/ATP transport, though the physiological relevance of this regulation remains unclear. We questioned whether myocardial ischemia provides a pathological setting in which PCoA regulation of ADP/ATP transport would be beneficial, and secondly, whether the chronically elevated lipid content within the diabetic heart could make mitochondria less sensitive to the effects of PCoA. PCoA acutely decreased ADP‐stimulated state 3 respiration and increased the apparent Km for ADP twofold. The half maximal inhibitory concentration (IC50) of PCoA in control mitochondria was 22 µM. This inhibitory effect of PCoA on respiration was blunted in diabetic mitochondria, with no significant difference in the Km for ADP in the presence of PCoA, and an increase in the IC50 to 32 µM PCoA. The competitive inhibition by PCoA was localised to the phosphorylation apparatus, particularly the ADP/ATP carrier (AAC). During ischemia, the AAC imports ATP into the mitochondria, where it is hydrolysed by reversal of the ATP synthase, regenerating the membrane potential. Addition of PCoA dose‐dependently prevented this wasteful ATP hydrolysis for membrane repolarisation during ischemia, however, this beneficial effect was blunted in diabetic mitochondria. Finally, using 31P‐magnetic resonance spectroscopy we demonstrated that diabetic hearts lose ATP more rapidly during ischemia, with a threefold higher ATP decay rate compared with control hearts. In conclusion, PCoA plays a role in protecting mitochondrial energetics during ischemia, by preventing wasteful ATP hydrolysis. However, this beneficial effect is blunted in diabetes, contributing to the impaired energy metabolism seen during myocardial ischemia in the diabetic heart.
Collapse
Affiliation(s)
- M Kerr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - K M J H Dennis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - C A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - W Fuller
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - G Berridge
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - S Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - C L Aitken
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - C Lopez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - R Fischer
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - J J Miller
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Department of Physics, University of Oxford, Oxford, UK.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - K Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - D J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - L C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Cheresh P, Kim SJ, Jablonski R, Watanabe S, Lu Z, Chi M, Helmin KA, Gius D, Budinger GRS, Kamp DW. SIRT3 Overexpression Ameliorates Asbestos-Induced Pulmonary Fibrosis, mt-DNA Damage, and Lung Fibrogenic Monocyte Recruitment. Int J Mol Sci 2021; 22:6856. [PMID: 34202229 PMCID: PMC8268084 DOI: 10.3390/ijms22136856] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 01/28/2023] Open
Abstract
Alveolar epithelial cell (AEC) mitochondrial (mt) DNA damage and fibrotic monocyte-derived alveolar macrophages (Mo-AMs) are implicated in the pathobiology of pulmonary fibrosis. We showed that sirtuin 3 (SIRT3), a mitochondrial protein regulating cell fate and aging, is deficient in the AECs of idiopathic pulmonary fibrosis (IPF) patients and that asbestos- and bleomycin-induced lung fibrosis is augmented in Sirt3 knockout (Sirt3-/-) mice associated with AEC mtDNA damage and intrinsic apoptosis. We determined whether whole body transgenic SIRT3 overexpression (Sirt3Tg) protects mice from asbestos-induced pulmonary fibrosis by mitigating lung mtDNA damage and Mo-AM recruitment. Crocidolite asbestos (100 µg/50 µL) or control was instilled intratracheally in C57Bl6 (Wild-Type) mice or Sirt3Tg mice, and at 21 d lung fibrosis (histology, fibrosis score, Sircol assay) and lung Mo-AMs (flow cytometry) were assessed. Compared to controls, Sirt3Tg mice were protected from asbestos-induced pulmonary fibrosis and had diminished lung mtDNA damage and Mo-AM recruitment. Further, pharmacologic SIRT3 inducers (i.e., resveratrol, viniferin, and honokiol) each diminish oxidant-induced AEC mtDNA damage in vitro and, in the case of honokiol, protection occurs in a SIRT3-dependent manner. We reason that SIRT3 preservation of AEC mtDNA is a novel therapeutic focus for managing patients with IPF and other types of pulmonary fibrosis.
Collapse
Affiliation(s)
- Paul Cheresh
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Seok-Jo Kim
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Renea Jablonski
- Section of Pulmonary and Critical Care, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Satoshi Watanabe
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Ziyan Lu
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Monica Chi
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - Kathryn A. Helmin
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - David Gius
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - G. R. Scott Budinger
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| | - David W. Kamp
- Jesse Brown VA Medical Center, Division of Pulmonary & Critical Care Medicine, Chicago, IL 60612, USA; (P.C.); (S.-J.K.); (Z.L.); (G.R.S.B.)
- Department of Medicine, Feinberg School of Medicine, Pulmonary and Critical Care Medicine, Northwestern University, Simpson & Querrey Biomedical Research Center 5-303, 303 E Superior St., Chicago, IL 60611, USA; (S.W.); (M.C.); (K.A.H.)
| |
Collapse
|
16
|
Resveratrol Prevents Right Ventricle Dysfunction, Calcium Mishandling, and Energetic Failure via SIRT3 Stimulation in Pulmonary Arterial Hypertension. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9912434. [PMID: 34239697 PMCID: PMC8238598 DOI: 10.1155/2021/9912434] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary vessel remodeling; however, its severity and impact on survival depend on right ventricular (RV) failure. Resveratrol (RES), a polyphenol found in red wine, exhibits cardioprotective effects on RV dysfunction in PAH. However, most literature has focused on RES protective effect on lung vasculature; recent finding indicates that RES has a cardioprotective effect independent of pulmonary arterial pressure on RV dysfunction, although the underlying mechanism in RV has not been determined. Therefore, this study is aimed at evaluating sirtuin-3 (SIRT3) modulation by RES in RV using a monocrotaline- (MC-) induced PAH rat model. Myocyte function was evaluated by confocal microscopy as cell contractility, calcium signaling, and mitochondrial membrane potential (ΔΨm); cell energetics was assessed by high-resolution respirometry, and western blot and immunoprecipitation evaluated posttranslational modifications. PAH significantly affects mitochondrial function in RV; PAH is prone to mitochondrial permeability transition pore (mPTP) opening, thus decreasing the mitochondrial membrane potential. The compromised cellular energetics affects cardiomyocyte function by decreasing sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) activity and delaying myofilament unbinding, disrupting cell relaxation. RES partially protects mitochondrial integrity by deacetylating cyclophilin-D, a critical component of the mPTP, increasing SIRT3 expression and activity and preventing mPTP opening. The preserved energetic capability rescues cell relaxation by maintaining SERCA activity. Avoiding Ca2+ transient and cell contractility mismatch by preserving mitochondrial function describes, for the first time, impairment in excitation-contraction-energetics coupling in RV failure. These results highlight the importance of mitochondrial energetics and mPTP in PAH.
Collapse
|
17
|
Jayakumari NR, Rajendran RS, Sivasailam A, Parambil ST, Reghuvaran AC, Sreelatha HV, Gopala S. Honokiol regulates mitochondrial substrate utilization and cellular fatty acid metabolism in diabetic mice heart. Eur J Pharmacol 2021; 896:173918. [PMID: 33529726 DOI: 10.1016/j.ejphar.2021.173918] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/13/2021] [Accepted: 01/26/2021] [Indexed: 01/06/2023]
Abstract
Type 2 diabetes mellitus is strongly associated with cardiac mitochondrial dysfunction, which is one of the main reasons for cardiovascular diseases. Among the mitochondrial metabolic changes, fatty acid metabolism is of great importance as cardiac tissues depend primarily on fatty acids. Honokiol, a constituent of Magnolia tree bark extract, is reported to strongly influence cardiac mitochondrial functions, via various mechanisms. The current study showed that honokiol decreased fatty acid-mediated complex I respiration and increased carbohydrate-mediated complex I and II respiration in diabetic C57BL/6 mice cardiac mitochondria. It was also found that honokiol treatment decreased expression of Cluster of Differentiation 36, AMP-activated kinases and nuclear transcription factors like, Peroxisome proliferator-activated receptor γ co-activator 1α/β and Peroxisome proliferator-activated receptor α, surrogating the evidence of decreased fatty acid-mediated complex I respiration. Honokiol treatment also reduced the levels of mitochondrial acetylated proteins, suggesting the possible action of honokiol via acetylation/deacetylation mechanism of regulation of protein functions in diabetic mitochondria. The antioxidant effect of honokiol is evidenced by the augmented expression of Manganese super oxide dismutase. In conclusion, honokiol imparts beneficial effect on diabetic cardiac mitochondria by decreasing the oxidant burden via regulating mitochondrial fatty acid respiration and expression of oxidant response factors.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Acetylation
- Animals
- Antioxidants/pharmacology
- Biphenyl Compounds/pharmacology
- CD36 Antigens/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Energy Metabolism/drug effects
- Fatty Acids/metabolism
- Lignans/pharmacology
- Male
- Mice, Inbred C57BL
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- NF-E2-Related Factor 2/metabolism
- Oxidative Stress/drug effects
- PPAR alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Streptozocin
- Superoxide Dismutase/metabolism
- Mice
Collapse
Affiliation(s)
- Nandini Ravikumar Jayakumari
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Raji Sasikala Rajendran
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Ashok Sivasailam
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Sulfath Thottungal Parambil
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Anand Chellappan Reghuvaran
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Harikrishnan Vijayakumar Sreelatha
- Division of Laboratory Animal Sciences, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
18
|
Zhou YQ, Mei W, Tian XB, Tian YK, Liu DQ, Ye DW. The therapeutic potential of Nrf2 inducers in chronic pain: Evidence from preclinical studies. Pharmacol Ther 2021; 225:107846. [PMID: 33819559 DOI: 10.1016/j.pharmthera.2021.107846] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Chronic pain remains an enormous health problem affecting approximatively 30% of the world's population. Opioids as the first line analgesics often leads to undesirable side effects when used long term. Therefore, novel therapeutic targets are urgently needed to the development of more efficacious analgesics. Substantial evidence indicates that excessive reactive oxygen species (ROS) are extremely important to the development of chronic pain. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master transcription factor regulating endogenous antioxidant defense. Emerging evidence suggests that Nrf2 and its downstream effectors are implicated in chronic inflammatory and neuropathic pain. Notably, controversial results have been reported regarding the expression of Nrf2 and its downstream targets in peripheral and central regions involved in pain transmission. However, our recent studies and results from other laboratories demonstrate that Nrf2 inducers exert potent analgesic effects in various murine models of chronic pain. In this review, we summarized and discussed the preclinical evidence demonstrating the therapeutic potential of Nrf2 inducers in chronic pain. These evidence indicates that Nrf2 activation are beneficial in chronic pain mostly by alleviating ROS-associated pathological processes. Overall, Nrf2-based therapy for chronic pain is an area with great promise, but more research regarding its detailed mechanisms is warranted.
Collapse
Affiliation(s)
- Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Mei
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xue-Bi Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Ke Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai-Qiang Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Da-Wei Ye
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University; Tongji Shanxi Hospital, Tongji Medical College, Huazhong University of Science and Technology, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|