1
|
Haque MM, Kuppusamy P, Melemedjian OK. Disruption of mitochondrial pyruvate oxidation in dorsal root ganglia drives persistent nociceptive sensitization and causes pervasive transcriptomic alterations. Pain 2024; 165:1531-1549. [PMID: 38285538 PMCID: PMC11189764 DOI: 10.1097/j.pain.0000000000003158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 01/31/2024]
Abstract
ABSTRACT Metabolism is inextricably linked to every aspect of cellular function. In addition to energy production and biosynthesis, metabolism plays a crucial role in regulating signal transduction and gene expression. Altered metabolic states have been shown to maintain aberrant signaling and transcription, contributing to diseases like cancer, cardiovascular disease, and neurodegeneration. Metabolic gene polymorphisms and defects are also associated with chronic pain conditions, as are increased levels of nerve growth factor (NGF). However, the mechanisms by which NGF may modulate sensory neuron metabolism remain unclear. This study demonstrated that intraplantar NGF injection reprograms sensory neuron metabolism. Nerve growth factor suppressed mitochondrial pyruvate oxidation and enhanced lactate extrusion, requiring 24 hours to increase lactate dehydrogenase A and pyruvate dehydrogenase kinase 1 (PDHK1) expression. Inhibiting these metabolic enzymes reversed NGF-mediated effects. Remarkably, directly disrupting mitochondrial pyruvate oxidation induced severe, persistent allodynia, implicating this metabolic dysfunction in chronic pain. Nanopore long-read sequencing of poly(A) mRNA uncovered extensive transcriptomic changes upon metabolic disruption, including altered gene expression, splicing, and poly(A) tail lengths. By linking metabolic disturbance of dorsal root ganglia to transcriptome reprogramming, this study enhances our understanding of the mechanisms underlying persistent nociceptive sensitization. These findings imply that impaired mitochondrial pyruvate oxidation may drive chronic pain, possibly by impacting transcriptomic regulation. Exploring these metabolite-driven mechanisms further might reveal novel therapeutic targets for intractable pain.
Collapse
Affiliation(s)
- Md Mamunul Haque
- Deptartmen of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Panjamurthy Kuppusamy
- Deptartmen of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Ohannes K. Melemedjian
- Deptartmen of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
- UM Center to Advance Chronic Pain Research, Baltimore, MD, United States
- UM Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
2
|
De Marco O, Gambardella J, Bianco A, Fiordelisi A, Cerasuolo FA, Buonaiuto A, Avvisato R, Capuano I, Amicone M, Di Risi T, Riccio E, Spinelli L, Pisani A, Iaccarino G, Sorriento D. Cardiopulmonary determinants of reduced exercise tolerance in Fabry disease. Front Cardiovasc Med 2024; 11:1396996. [PMID: 38756750 PMCID: PMC11096481 DOI: 10.3389/fcvm.2024.1396996] [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: 03/06/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024] Open
Abstract
Fabry disease (FD), also known as Anderson-Fabry disease, is a hereditary disorder of glycosphingolipid metabolism, caused by a deficiency of the lysosomal alpha-galactosidase A enzyme. This causes a progressive accumulation of glycosphingolipids in tissues and organs which represents the main pathogenetic mechanism of FD. The disease is progressive and multisystemic and is characterized by early symptoms and late complications (renal, cardiac and neurological dysfunction). Fatigue and exercise intolerance are early common symptoms in FD patients but the specific causes are still to be defined. In this narrative review, we deal with the contribution of cardiac and pulmonary dysfunctions in determining fatigue and exercise intolerance in FD patients.
Collapse
Affiliation(s)
- Oriana De Marco
- Department of Public Health, Federico II University, Naples, Italy
| | - Jessica Gambardella
- Centro Interdipartimentale di Ricerca in Ipertensione Arteriosa e Patologie Associate, Federico II University of Naples, Naples, Italy
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Antonio Bianco
- Department of Public Health, Federico II University, Naples, Italy
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | | | - Antonietta Buonaiuto
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Roberta Avvisato
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Ivana Capuano
- Department of Public Health, Federico II University, Naples, Italy
| | - Maria Amicone
- Department of Public Health, Federico II University, Naples, Italy
| | - Teodolinda Di Risi
- Department of Public Health, Federico II University, Naples, Italy
- CEINGE - Biotecnologie Avanzate, Naples, Italy
| | - Eleonora Riccio
- Department of Public Health, Federico II University, Naples, Italy
| | - Letizia Spinelli
- Centro Interdipartimentale di Ricerca in Ipertensione Arteriosa e Patologie Associate, Federico II University of Naples, Naples, Italy
| | - Antonio Pisani
- Department of Public Health, Federico II University, Naples, Italy
- Centro Interdipartimentale di Ricerca in Ipertensione Arteriosa e Patologie Associate, Federico II University of Naples, Naples, Italy
| | - Guido Iaccarino
- Centro Interdipartimentale di Ricerca in Ipertensione Arteriosa e Patologie Associate, Federico II University of Naples, Naples, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Daniela Sorriento
- Centro Interdipartimentale di Ricerca in Ipertensione Arteriosa e Patologie Associate, Federico II University of Naples, Naples, Italy
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
3
|
Meng S, Yu Y, Yu S, Zhu S, Shi M, Xiang M, Ma H. Advances in Metabolic Remodeling and Intervention Strategies in Heart Failure. J Cardiovasc Transl Res 2024; 17:36-55. [PMID: 37843752 DOI: 10.1007/s12265-023-10443-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
The heart is the most energy-demanding organ throughout the whole body. Perturbations or failure in energy metabolism contributes to heart failure (HF), which represents the advanced stage of various heart diseases. The poor prognosis and huge economic burden associated with HF underscore the high unmet need to explore novel therapies targeting metabolic modulators beyond conventional approaches focused on neurohormonal and hemodynamic regulators. Emerging evidence suggests that alterations in metabolic substrate reliance, metabolic pathways, metabolic by-products, and energy production collectively regulate the occurrence and progression of HF. In this review, we provide an overview of cardiac metabolic remodeling, encompassing the utilization of free fatty acids, glucose metabolism, ketone bodies, and branched-chain amino acids both in the physiological condition and heart failure. Most importantly, the latest advances in pharmacological interventions are discussed as a promising therapeutic approach to restore cardiac function, drawing insights from recent basic research, preclinical and clinical studies.
Collapse
Affiliation(s)
- Simin Meng
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Yi Yu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Shiyu Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mengjia Shi
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
4
|
Liu Y, Lin X, Hao Z, Yu M, Tang Y, Teng X, Sun W, Kang L. Cadmium exposure caused cardiotoxicity in common carps (Cyprinus carpio L.): miR-9-5p, oxidative stress, energetic impairment, mitochondrial division/fusion imbalance, inflammation, and autophagy. FISH & SHELLFISH IMMUNOLOGY 2023; 138:108853. [PMID: 37245677 DOI: 10.1016/j.fsi.2023.108853] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 05/30/2023]
Abstract
Cadmium (Cd), a toxic heavy metal pollutant, is a threat to human and eatable fish health. Common carps are widely cultivated and eaten by humans. However, there are no reports about Cd-damaged common carp hearts. Our experiment attempted to investigate the cardiotoxicity of Cd to common carps by establishing a common carp Cd exposure model. Our results showed that Cd injured hearts. Moreover, Cd treatment induced autophagy via miR-9-5p/Sirt1/mTOR/ULK1 pathway. Cd exposure caused oxidant/antioxidant imbalance and oxidative stress; and led to energetic impairment. Energetic impairment partook in oxidative stress-mediated autophagy through AMPK/mTOR/ULK1 pathway. Furthermore, Cd caused mitochondrial division/fusion imbalance and resulted in inflammatory injury via NF-κB-COX-2-PTGEs and NF-κB-COX-2-TNF-α pathways. Oxidative stress mediated mitochondrial division/fusion imbalance, further induced inflammation and autophagy via OPA1/NF-κB-COX-2-TNF-α-Beclin1 and OPA1/NF-κB-COX-2-TNF-α/P62 pathways under Cd treatment. Taken together, miR-9-5p, oxidative stress, energetic impairment, mitochondrial division/fusion imbalance, inflammation, and autophagy participated in the mechanism of Cd-cardiotoxicity to common carps. Our study revealed harmful effect of Cd on hearts, and provided new information for researches of environmental pollutant toxicity.
Collapse
Affiliation(s)
- Yuhao Liu
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - Xu Lin
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - Zhiyu Hao
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - Meijing Yu
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - You Tang
- Electrical and Information Engineering College, JiLin Agricultural Science and Technology University, Jilin, 132101, PR China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China.
| | - Wei Sun
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China.
| | - Lu Kang
- Institute of Agricultural Quality Standards and Testing Technology, Xinjiang Academy of Agricultural Sciences, Urumqi, 830091, PR China.
| |
Collapse
|
5
|
Dhar A, Venkadakrishnan J, Roy U, Vedam S, Lalwani N, Ramos KS, Pandita TK, Bhat A. A comprehensive review of the novel therapeutic targets for the treatment of diabetic cardiomyopathy. Ther Adv Cardiovasc Dis 2023; 17:17539447231210170. [PMID: 38069578 PMCID: PMC10710750 DOI: 10.1177/17539447231210170] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 10/09/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is characterized by structural and functional abnormalities in the myocardium affecting people with diabetes. Treatment of DCM focuses on glucose control, blood pressure management, lipid-lowering, and lifestyle changes. Due to limited therapeutic options, DCM remains a significant cause of morbidity and mortality in patients with diabetes, thus emphasizing the need to develop new therapeutic strategies. Ongoing research is aimed at understanding the underlying molecular mechanism(s) involved in the development and progression of DCM, including oxidative stress, inflammation, and metabolic dysregulation. The goal is to develope innovative pharmaceutical therapeutics, offering significant improvements in the clinical management of DCM. Some of these approaches include the effective targeting of impaired insulin signaling, cardiac stiffness, glucotoxicity, lipotoxicity, inflammation, oxidative stress, cardiac hypertrophy, and fibrosis. This review focuses on the latest developments in understanding the underlying causes of DCM and the therapeutic landscape of DCM treatment.
Collapse
Affiliation(s)
- Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | | | - Utsa Roy
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Sahithi Vedam
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Nikita Lalwani
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana, India
| | - Kenneth S. Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Tej K. Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir (UT) 184311, India
| |
Collapse
|
6
|
Aczel D, Gyorgy B, Bakonyi P, BukhAri R, Pinho R, Boldogh I, Yaodong G, Radak Z. The Systemic Effects of Exercise on the Systemic Effects of Alzheimer's Disease. Antioxidants (Basel) 2022; 11:antiox11051028. [PMID: 35624892 PMCID: PMC9137920 DOI: 10.3390/antiox11051028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive degenerative disorder and a leading cause of dementia in the elderly. The etiology of AD is multifactorial, including an increased oxidative state, deposition of amyloid plaques, and neurofibrillary tangles of the tau protein. The formation of amyloid plaques is considered one of the first signs of the illness, but only in the central nervous system (CNS). Interestingly, results indicate that AD is not just localized in the brain but is also found in organs distant from the brain, such as the cardiovascular system, gut microbiome, liver, testes, and kidney. These observations make AD a complex systemic disorder. Still, no effective medications have been found, but regular physical activity has been considered to have a positive impact on this challenging disease. While several articles have been published on the benefits of physical activity on AD development in the CNS, its peripheral effects have not been discussed in detail. The provocative question arising is the following: is it possible that the beneficial effects of regular exercise on AD are due to the systemic impact of training, rather than just the effects of exercise on the brain? If so, does this mean that the level of fitness of these peripheral organs can directly or indirectly influence the incidence or progress of AD? Therefore, the present paper aims to summarize the systemic effects of both regular exercise and AD and point out how common exercise-induced adaptation via peripheral organs can decrease the incidence of AD or attenuate the progress of AD.
Collapse
Affiliation(s)
- Dora Aczel
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - Bernadett Gyorgy
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - Peter Bakonyi
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - RehAn BukhAri
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - Ricardo Pinho
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil;
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA;
| | - Gu Yaodong
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China;
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
- Correspondence: ; Tel.: +36-1-3565764; Fax: +36-1-3566337
| |
Collapse
|
7
|
Murphy J, Le TNV, Fedorova J, Yang Y, Krause-Hauch M, Davitt K, Zoungrana LI, Fatmi MK, Lesnefsky EJ, Li J, Ren D. The Cardiac Dysfunction Caused by Metabolic Alterations in Alzheimer's Disease. Front Cardiovasc Med 2022; 9:850538. [PMID: 35274014 PMCID: PMC8902161 DOI: 10.3389/fcvm.2022.850538] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
A progressive defect in the energy generation pathway is implicated in multiple aging-related diseases, including cardiovascular conditions and Alzheimer's Disease (AD). However, evidence of the pathogenesis of cardiac dysfunction in AD and the associations between the two organ diseases need further elucidation. This study aims to characterize cellular defects resulting in decreased cardiac function in AD-model. 5XFAD mice, a strain expressing five mutations in human APP and PS1 that shows robust Aβ production with visible plaques at 2 months and were used in this study as a model of AD. 5XFAD mice and wild-type (WT) counterparts were subjected to echocardiography at 2-, 4-, and 6-month, and 5XFAD had a significant reduction in cardiac fractional shortening and ejection fraction compared to WT. Additionally, 5XFAD mice had decreased observed electrical signals demonstrated as decreased R, P, T wave amplitudes. In isolated cardiomyocytes, 5XFAD mice showed decreased fraction shortening, rate of shortening, as well as the degree of transient calcium influx. To reveal the mechanism by which AD leads to cardiac systolic dysfunction, the immunoblotting analysis showed increased activation of AMP-activated protein kinase (AMPK) in 5XFAD left ventricular and brain tissue, indicating altered energy metabolism. Mito Stress Assays examining mitochondrial function revealed decreased basal and maximal oxygen consumption rate, as well as defective pyruvate dehydrogenase activity in the 5XFAD heart and brain. Cellular inflammation was provoked in the 5XFAD heart and brain marked by the increase of reactive oxygen species accumulation and upregulation of inflammatory mediator activities. Finally, AD pathological phenotype with increased deposition of Aβ and defective cognitive function was observed in 6-month 5XFAD mice. In addition, elevated fibrosis was observed in the 6-month 5XFAD heart. The results implicated that AD led to defective mitochondrial function, and increased inflammation which caused the decrease in contractility of the heart.
Collapse
Affiliation(s)
- Jiayuan Murphy
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Tran Ngoc Van Le
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Julia Fedorova
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Yi Yang
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Meredith Krause-Hauch
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Kayla Davitt
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Linda Ines Zoungrana
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mohammad Kasim Fatmi
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Edward J. Lesnefsky
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
- Cardiology Section, Medical Service, Richmond Department of Veterans Affairs Medical Center, Richmond, VA, United States
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Di Ren
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
8
|
Tognola C, Alessandro M, Milani M, Cartella I, Tavecchia G, Grasso E, Sun J, Giannattasio C. Nutraceuticals in Chronic Coronary Syndromes: Preclinical Data and Translational Experiences. High Blood Press Cardiovasc Prev 2021; 28:13-25. [PMID: 33125662 PMCID: PMC7864844 DOI: 10.1007/s40292-020-00416-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Non-pharmacological treatments have always been considered important in the management of Chronic Coronary Syndromes. Nutraceuticals ("Nutrition" + "Pharmaceutical") could fall both under the definition of non-pharmacological treatment and pharmacological one or, probably more correctly, in the middle of these two kinds of therapies. However, the word "nutraceuticals" never appears in the latest guidelines on this issue. This is probably determined by the fact that evidences on this topic are scarce and most of the published articles are based on preclinical data while translational experiences are available only for some molecules. In this review we will focus on nutraceutical strategies that act on the ischemic myocardium itself and not only on the cardiovascular risk factors. As demonstrated by the important number of papers published in recent years, this is an evolving topic and evaluated substances principally act on two mechanisms (cardiac energetics and ischemia-reperfusion damage) that will be also reviewed.
Collapse
Affiliation(s)
- Chiara Tognola
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy
| | - Maloberti Alessandro
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy.
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy.
| | - Martina Milani
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy
| | - Iside Cartella
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy
| | - Giovanni Tavecchia
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy
| | - Enzo Grasso
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy
| | - Jinwey Sun
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy
| | - Cristina Giannattasio
- Cardiologia IV, Dipartimento A. De Gasperis, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, Milan, Italy
| |
Collapse
|
9
|
Tona F, Boscaro M, Barbot M, Maritan L, Famoso G, Dal Lin C, Montisci R, Fallo F, Iliceto S, Scaroni C. New insights to the potential mechanisms driving coronary flow reserve impairment in Cushing's syndrome: A pilot noninvasive study by transthoracic Doppler echocardiography. Microvasc Res 2020; 128:103940. [DOI: 10.1016/j.mvr.2019.103940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 01/06/2023]
|
10
|
Zhao Y, Nie S, Yi M, Wu N, Wang W, Zhang Z, Yao Y, Wang D. UPLC-QTOF/MS-based metabolomics analysis of plasma reveals an effect of Xue-Fu-Zhu-Yu capsules on blood-stasis syndrome in CHD rats. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:111908. [PMID: 31029757 DOI: 10.1016/j.jep.2019.111908] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Blood-stasis syndrome (BSS) is a specific ZHENG type of coronary heart disease (CHD) in traditional Chinese medicine (TCM). The Xue-Fu-Zhu-Yu (XFZY) decoction is a common herbal formula that has been used for several centuries to treat BSS, but its mechanism has not been thoroughly elucidated to date. AIM OF THE STUDY In this study, serum lipid, blood haemorheology and metabolomics analyses were performed to depict a complete profile of XFZY capsules for the treatment of CHD with BSS and to reveal the potential mechanism of the XFZY capsules. MATERIALS AND METHODS A rat model of CHD with BSS was generated by combining a high-fat diet (HFD) with a left anterior descending coronary artery (LAD) ligation. After four weeks of treatment with XFZY capsules or simvastatin pills, an echocardiography was performed for a therapeutic evaluation. Blood samples and heart tissues were then collected for further analyses. A UPLC-QTOF/MS-based metabolomics analysis of the plasma was performed, and all metabolic features were fit by PCA and OPLS-DA pattern for the biomarker screen. The identified biomarkers were later implemented into a metabolic pathway analysis. Furthermore, we used qRT-PCR and Western blot analyses to verify the treatment effects of the XFZY capsules. RESULTS A total of 49 metabolites (VIP>1.0, p < 0.05, RSD%<20%) were identified in the Model rats, and 27 metabolites (VIP>1.0, p < 0.05, RSD%<20%) were identified in the XFZY-H rats. The results of the pathway analysis indicated that the XFZY capsules treated CHD primarily by regulating cardiac energy, phospholipid, polyunsaturated fatty acid (PUFA) and amino acid metabolism. In addition, blood viscosity and serum lipid assays suggested that XFZY capsules could decrease serum triglycerides, total cholesterol, low-density lipoprotein cholesterol and whole blood viscosity at a low shear rate. CONCLUSION This study demonstrated that the XFZY capsule effectively decreases serum lipids and whole blood viscosity in CHD with BSS. The underlying metabolic mechanism mainly included improving cardiac energy supply, reducing phospholipid peroxide, maintaining the PUFA metabolic balance and regulating amino acid metabolism.
Collapse
Affiliation(s)
- Yuhang Zhao
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, Hunan, 410008, China.
| | - Shanshan Nie
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, Hunan, 410008, China.
| | - Min Yi
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, Hunan, 410008, China.
| | - Ning Wu
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, Hunan, 410008, China.
| | - Wenbo Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, Hunan, 410008, China.
| | - Zheyu Zhang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Ye Yao
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, Hunan, 410008, China.
| | - Dongsheng Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
11
|
Pierce JD, Mahoney DE, Hiebert JB, Thimmesch AR, Diaz FJ, Smith C, Shen Q, Mudaranthakam DP, Clancy RL. Study protocol, randomized controlled trial: reducing symptom burden in patients with heart failure with preserved ejection fraction using ubiquinol and/or D-ribose. BMC Cardiovasc Disord 2018; 18:57. [PMID: 29606104 PMCID: PMC5879598 DOI: 10.1186/s12872-018-0796-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/20/2018] [Indexed: 12/15/2022] Open
Abstract
Background Heart failure (HF), the leading cause of morbidity and mortality in the US, affects 6.6 million adults with an estimated additional 3 million people by 2030. More than 50% of HF patients have heart failure with preserved left ventricular ejection fraction (HFpEF). These patients have impaired cardiac muscle relaxation and diastolic filling, which investigators have associated with cellular energetic impairment. Patients with HFpEF experience symptoms of: (1) fatigue; (2) shortness of breath; and (3) swelling (edema) of the lower extremities. However, current HF guidelines offer no effective treatment to address these underlying pathophysiologic mechanisms. Thus, we propose a biobehavioral symptom science study using ubiquinol and D-ribose (therapeutic interventions) to target mitochondrial bioenergetics to reduce the complex symptoms experienced by patients with HFpEF. Methods Using a randomized, double-blind, placebo-controlled design, the overall objective is to determine if administering ubiquinol and/or D-ribose to HFpEF patients for 12 weeks would decrease the severity of their complex symptoms and improve their cardiac function. The measures used to assess patients’ perceptions of their health status and level of vigor (energy) will be the Kansas City Cardiomyopathy Questionnaire (KCCQ) and Vigor subscale of the Profile of Mood States. The 6-min walk test will be used to test exercise tolerance. Left ventricular diastolic function will be assessed using innovative advanced echocardiography software called speckle tracking. We will measure B-type natriuretic peptides (secreted from ventricles in HF) and lactate/ATP ratio (measure of cellular energetics). Discussions Ubiquinol (active form of Coenzyme Q10) and D-ribose are two potential treatments that can positively affect cellular energetic impairment, the major underlying mechanism of HFpEF. Ubiquinol, the reduced form of CoQ10, is more effective in adults over the age of 50. In patients with HFpEF, mitochondrial deficiency of ubiquinol results in decreased adenosine triphosphate (ATP) synthesis and reduced scavenging of reactive oxygen species. D-ribose is a substrate required for ATP synthesis and when administered has been shown to improve impaired myocardial bioenergetics. Therefore, if the biological underpinning of deficient mitochondrial ATP in HFpEF is not addressed, patients will suffer major symptoms including lack of energy, fatigue, exertional dyspnea, and exercise intolerance. Trial registration ClinicalTrials.gov Identifier: NCT03133793; Data of Registration: April 28, 2017.
Collapse
Affiliation(s)
- Janet D Pierce
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Diane E Mahoney
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - John B Hiebert
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Amanda R Thimmesch
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Francisco J Diaz
- Department of Biostatistics, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Carol Smith
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Qiuhua Shen
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Dinesh Pal Mudaranthakam
- Department of Biostatistics, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Richard L Clancy
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| |
Collapse
|
12
|
Myocardial metabolic alterations in mice with diet-induced atherosclerosis: linking sulfur amino acid and lipid metabolism. Sci Rep 2017; 7:13597. [PMID: 29051579 PMCID: PMC5648757 DOI: 10.1038/s41598-017-13991-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular disease (CVD), but the effect of diet on the atherosclerotic heart’s metabolism is unclear. We used an integrated metabolomics and lipidomics approach to evaluate metabolic perturbations in heart and serum from mice fed an atherogenic diet (AD) for 8, 16, and 25 weeks. Nuclear magnetic resonance (NMR)-based metabolomics revealed significant changes in sulfur amino acid (SAA) and lipid metabolism in heart from AD mice compared with heart from normal diet mice. Higher SAA levels in AD mice were quantitatively verified using liquid chromatography-mass spectrometry (LC/MS). Lipidomic profiling revealed that fatty acid and triglyceride (TG) levels in the AD group were altered depending on the degree of unsaturation. Additionally, levels of SCD1, SREBP-1, and PPARγ were reduced in AD mice after 25 weeks, while levels of reactive oxygen species were elevated. The results suggest that a long-term AD leads to SAA metabolism dysregulation and increased oxidative stress in the heart, causing SCD1 activity suppression and accumulation of toxic TGs with a low degree of unsaturation. These findings demonstrate that the SAA metabolic pathway is a promising therapeutic target for CVD treatment and that metabolomics can be used to investigate the metabolic signature of atherosclerosis.
Collapse
|
13
|
Makrecka-Kuka M, Volska K, Antone U, Vilskersts R, Grinberga S, Bandere D, Liepinsh E, Dambrova M. Trimethylamine N-oxide impairs pyruvate and fatty acid oxidation in cardiac mitochondria. Toxicol Lett 2017; 267:32-38. [DOI: 10.1016/j.toxlet.2016.12.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/16/2016] [Accepted: 12/29/2016] [Indexed: 02/08/2023]
|
14
|
George CH, Mitchell AN, Preece R, Bannister ML, Yousef Z. Pleiotropic mechanisms of action of perhexiline in heart failure. Expert Opin Ther Pat 2016; 26:1049-59. [PMID: 27455171 DOI: 10.1080/13543776.2016.1211111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The re-purposing of the anti-anginal drug perhexiline (PHX) has resulted in symptomatic improvements in heart failure (HF) patients. The inhibition of carnitine palmitoyltransferase-1 (CPT-1) has been proposed as the primary mechanism underlying the therapeutic benefit of PHX. This hypothesis is contentious. AREAS COVERED We reviewed the primary literature and patent landscape of PHX from its initial development in the 1960s through to its emergence as a drug beneficial for HF. We focused on its physico-chemistry, molecular targets, tissue accumulation and clinical dosing. EXPERT OPINION Dogma that the beneficial effects of PHX are due primarily to potent myocardial CPT-1 inhibition is not supported by the literature and all available evidence point to it being extremely unlikely that the major effects of PHX occur via this mechanism. In vivo PHX is much more likely to be an inhibitor of surface membrane ion channels and also to have effects on other components of cellular metabolism and reactive oxygen species (ROS) generation across the cardiovascular system. However, the possibility that minor effects of PHX on CPT-1 underpin disproportionately large effects on myocardial function cannot be entirely excluded, especially given the massive accumulation of the drug in heart tissue.
Collapse
Affiliation(s)
- Christopher H George
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Alice N Mitchell
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Ryan Preece
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Mark L Bannister
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Zaheer Yousef
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| |
Collapse
|
15
|
|
16
|
Loudon BL, Noordali H, Gollop ND, Frenneaux MP, Madhani M. Present and future pharmacotherapeutic agents in heart failure: an evolving paradigm. Br J Pharmacol 2016; 173:1911-24. [PMID: 26993743 PMCID: PMC4882493 DOI: 10.1111/bph.13480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/28/2016] [Accepted: 02/26/2016] [Indexed: 02/06/2023] Open
Abstract
Many conditions culminate in heart failure (HF), a multi‐organ systemic syndrome with an intrinsically poor prognosis. Pharmacotherapeutic agents that correct neurohormonal dysregulation and haemodynamic instability have occupied the forefront of developments within the treatment of HF in the past. Indeed, multiple trials aimed to validate these agents in the 1980s and early 1990s, resulting in a large and robust evidence‐base supporting their use clinically. An established treatment paradigm now exists for the treatment of HF with reduced ejection fraction (HFrEF), but there have been very few notable developments in recent years. HF remains a significant health concern with an increasing incidence as the population ages. We may indeed be entering the surgical era for HF treatment, but these therapies remain expensive and inaccessible to many. Newer pharmacotherapeutic agents are slowly emerging, many targeting alternative therapeutic pathways, but with mixed results. Metabolic modulation and manipulation of the nitrate/nitrite/nitric oxide pathway have shown promise and could provide the answers to fill the therapeutic gap between medical interventions and surgery, but further definitive trials are warranted. We review the significant evidence base behind the current medical treatments for HFrEF, the physiology of metabolic impairment in HF, and discuss two promising novel agents, perhexiline and nitrite.
Collapse
Affiliation(s)
- Brodie L Loudon
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Hannah Noordali
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nicholas D Gollop
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Michael P Frenneaux
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
17
|
Davogustto G, Taegtmeyer H. The changing landscape of cardiac metabolism. J Mol Cell Cardiol 2015; 84:129-32. [PMID: 25937535 DOI: 10.1016/j.yjmcc.2015.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/26/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Giovanni Davogustto
- Division of Cardiology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|