1
|
Williams LM, Cao S. Harnessing and delivering microbial metabolites as therapeutics via advanced pharmaceutical approaches. Pharmacol Ther 2024; 256:108605. [PMID: 38367866 PMCID: PMC10985132 DOI: 10.1016/j.pharmthera.2024.108605] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Abstract
Microbial metabolites have emerged as key players in the interplay between diet, the gut microbiome, and host health. Two major classes, short-chain fatty acids (SCFAs) and tryptophan (Trp) metabolites, are recognized to regulate inflammatory, immune, and metabolic responses within the host. Given that many human diseases are associated with dysbiosis of the gut microbiome and consequent reductions in microbial metabolite production, the administration of these metabolites represents a direct, multi-targeted treatment. While a multitude of preclinical studies showcase the therapeutic potential of both SCFAs and Trp metabolites, they often rely on high doses and frequent dosing regimens to achieve systemic effects, thereby constraining their clinical applicability. To address these limitations, a variety of pharmaceutical formulations approaches that enable targeted, delayed, and/or sustained microbial metabolite delivery have been developed. These approaches, including enteric encapsulations, esterification to dietary fiber, prodrugs, and nanoformulations, pave the way for the next generation of microbial metabolite-based therapeutics. In this review, we first provide an overview of the roles of microbial metabolites in maintaining host homeostasis and outline how compromised metabolite production contributes to the pathogenesis of inflammatory, metabolic, autoimmune, allergic, infectious, and cancerous diseases. Additionally, we explore the therapeutic potential of metabolites in these disease contexts. Then, we provide a comprehensive and up-to-date review of the pharmaceutical strategies that have been employed to enhance the therapeutic efficacy of microbial metabolites, with a focus on SCFAs and Trp metabolites.
Collapse
Affiliation(s)
- Lindsey M Williams
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Shijie Cao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
2
|
Liu T, Sun Z, Yang Z, Qiao X. Microbiota-derived short-chain fatty acids and modulation of host-derived peptides formation: Focused on host defense peptides. Biomed Pharmacother 2023; 162:114586. [PMID: 36989711 DOI: 10.1016/j.biopha.2023.114586] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The byproducts of bacterial fermentation known as short-chain fatty acids (SCFAs) are chemically comprised of a carboxylic acid component and a short hydrocarbon chain. Recent investigations have demonstrated that SCFAs can affect intestinal immunity by inducing endogenous host defense peptides (HDPs) and their beneficial effects on barrier integrity, gut health, energy supply, and inflammation. HDPs, which include defensins, cathelicidins, and C-type lectins, perform a significant function in innate immunity in gastrointestinal mucosal membranes. SCFAs have been demonstrated to stimulate HDP synthesis by intestinal epithelial cells via interactions with G protein-coupled receptor 43 (GPR43), activation of the Jun N-terminal kinase (JNK) and Mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways, and the cell growth pathways. Furthermore, SCFA butyrate has been demonstrated to enhance the number of HDPs released from macrophages. SCFAs promote monocyte-to-macrophage development and stimulate HDP synthesis in macrophages by inhibiting histone deacetylase (HDAC). Understanding the etiology of many common disorders might be facilitated by studies into the function of microbial metabolites, such as SCFAs, in the molecular regulatory processes of immune responses (e.g., HDP production). This review will focus on the current knowledge of the role and mechanism of microbiota-derived SCFAs in influencing the synthesis of host-derived peptides, particularly HDPs.
Collapse
|
3
|
Short-Chain Fatty Acids in Gut-Heart Axis: Their Role in the Pathology of Heart Failure. J Pers Med 2022; 12:jpm12111805. [PMID: 36579524 PMCID: PMC9695649 DOI: 10.3390/jpm12111805] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Heart failure (HF) is a syndrome with global clinical and socioeconomic burden worldwide owing to its poor prognosis. Accumulating evidence has implicated the possible contribution of gut microbiota-derived metabolites, short-chain fatty acids (SCFAs), on the pathology of a variety of diseases. The changes of SCFA concentration were reported to be observed in various cardiovascular diseases including HF in experimental animals and humans. HF causes hypoperfusion and/or congestion in the gut, which may lead to lowered production of SCFAs, possibly through the pathological changes of the gut microenvironment including microbiota composition. Recent studies suggest that SCFAs may play a significant role in the pathology of HF, possibly through an agonistic effect on G-protein-coupled receptors, histone deacetylases (HDACs) inhibition, restoration of mitochondrial function, amelioration of cardiac inflammatory response, its utilization as an energy source, and remote effect attributable to a protective effect on the other organs. Collectively, in the pathology of HF, SCFAs might play a significant role as a key mediator in the gut-heart axis. However, these possible mechanisms have not been entirely clarified and need further investigation.
Collapse
|
4
|
Copur S, Rossing P, Afsar B, Sag AA, Siriopol D, Kuwabara M, Ortiz A, Kanbay M. A primer on metabolic memory: why existing diabesity treatments fail. Clin Kidney J 2021; 14:756-767. [PMID: 34512957 PMCID: PMC8422888 DOI: 10.1093/ckj/sfaa143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 11/28/2022] Open
Abstract
Despite massive government and private sector investments into prevention of cardiovascular disease, diabetes mellitus and obesity, efforts have largely failed, and the burden of cost remains in the treatment of downstream morbidity and mortality, with overall stagnating outcomes. A new paradigm shift in the approach to these patients may explain why existing treatment strategies fail, and offer new treatment targets. This review aims to provide a clinician-centred primer on metabolic memory, defined as the sum of irreversible genetic, epigenetic, cellular and tissue-level alterations that occur with long-time exposure to metabolic derangements.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Baris Afsar
- Department of Internal Medicine, Division of Nephrology, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Alan A Sag
- Department of Radiology, Division of Vascular and Interventional Radiology, Duke University Medical Center, Durham, NC, USA
| | - Dimitrie Siriopol
- Nephrology Clinic, Dialysis and Renal Transplant Center, 'C.I. PARHON' University Hospital, 'Grigore T. Popa' University of Medicine, Iasi, Romania
| | | | - Alberto Ortiz
- School of Medicine, Dialysis Unit, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
5
|
Thiruvengadam M, Subramanian U, Venkidasamy B, Thirupathi P, Samynathan R, Shariati MA, Rebezov M, Chung IM, Rengasamy KRR. Emerging role of nutritional short-chain fatty acids (SCFAs) against cancer via modulation of hematopoiesis. Crit Rev Food Sci Nutr 2021; 63:827-844. [PMID: 34319824 DOI: 10.1080/10408398.2021.1954874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The understanding of gut microbiota has emerged as a significant frontier in development of strategies to maintain normal human body's homeostasis and preventing the disease development over the last decade. The composition of the gut microbiota influences the clinical benefit of immune checkpoints in patients with advanced cancer, but the mechanisms underlying this relationship are unclear. Cancer is among the leading causes of mortality worldwide. So far, there is no universal treatment for cancer and despite significant advances, a lot of improvement on cancer therapy is required. Owing to its role in preserving the host's health and maintaining cellular integrity, the human gut microbiome has recently drawn a lot of interest as a target for cancer treatment. Dietary fiber is fermented by the gut microbiota to generate short-chain fatty acids (SCFAs), such as acetate, butyrate, and propionate, which are physiologically active metabolites. SCFAs can modulate the pathophysiology of the tumor environment through various critical signaling pathways. In addition, SCFAs can bind to carcinogens and other toxic chemicals, thus facilitating their biotransformation and elimination through different excretory mechanisms. This review discusses the mechanisms of action of short-chain fatty acids in modulating hematopoiesis of various immune system cells and the resultant beneficial anti-cancer effects. It also provides future perspectives on cancer therapy.
Collapse
Affiliation(s)
- Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| | - Umadevi Subramanian
- Translational Research Platform for Veterinary Biologicals, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Baskar Venkidasamy
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, India
| | - Prabhu Thirupathi
- Translational Research Platform for Veterinary Biologicals, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | | | - Mohammad Ali Shariati
- Department of Technology of Food Products, K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), Moscow, Russian Federation
| | - Maksim Rebezov
- V M Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, Moscow, Russian Federation.,Prokhorov General Physics Institute of the Russian Academy of Science, Moscow, Russian Federation
| | - Ill-Min Chung
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| | - Kannan R R Rengasamy
- Green Biotechnologies Research Centre of Excellence, University of Limpopo, Mankweng, South Africa
| |
Collapse
|
6
|
Xie M, Cho GW, Kong Y, Li DL, Altamirano F, Luo X, Morales CR, Jiang N, Schiattarella GG, May HI, Medina J, Shelton J, Ferdous A, Gillette TG, Hill JA. Activation of Autophagic Flux Blunts Cardiac Ischemia/Reperfusion Injury. Circ Res 2021; 129:435-450. [PMID: 34111934 PMCID: PMC8317428 DOI: 10.1161/circresaha.120.318601] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/09/2021] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Min Xie
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Geoffrey W. Cho
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Yongli Kong
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Dan L. Li
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Xiang Luo
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Cyndi R. Morales
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Nan Jiang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Gabriele G. Schiattarella
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Herman I. May
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Jessica Medina
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - John Shelton
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Anwarul Ferdous
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| |
Collapse
|
7
|
Nudelman V, Zahalka MA, Nudelman A, Rephaeli A, Kessler-Icekson G. Cardioprotection by AN-7, a prodrug of the histone deacetylase inhibitor butyric acid: Selective activity in hypoxic cardiomyocytes and cardiofibroblasts. Eur J Pharmacol 2020; 882:173255. [PMID: 32553737 DOI: 10.1016/j.ejphar.2020.173255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/19/2020] [Accepted: 06/05/2020] [Indexed: 12/27/2022]
Abstract
The anticancer prodrug butyroyloxymethyl diethylphosphate (AN-7), upon metabolic hydrolysis, releases the histone deacetylase inhibitor butyric acid and imparts histone hyperacetylation. We have shown previously that AN-7 increases doxorubicin-induced cancer cell death and reduces doxorubicin toxicity and hypoxic damage to the heart and cardiomyocytes. The cardiofibroblasts remain unprotected against both insults. Herein we examined the selective effect of AN-7 on hypoxic cardiomyocytes and cardiofibroblasts and investigated mechanisms underlying the cell specific response. Hypoxic cardiomyocytes and cardiofibroblasts or H2O2-treated H9c2 cardiomyoblasts, were treated with AN-7 and cell damage and death were evaluated as well as cell signaling pathways and the expression levels of heme oxygenase-1 (HO-1). AN-7 diminished hypoxia-induced mitochondrial damage and cell death in hypoxic cardiomyocytes and reduced hydrogen peroxide damage in H9c2 cells while increasing cell injury and death in hypoxic cardiofibroblasts. In the cell line, AN-7 induced Akt and ERK survival pathway activation in a kinase-specific manner including phosphorylation of the respective downstream targets, GSK-3β and BAD. Hypoxic cardiomyocytes responded to AN-7 treatment by enhanced phosphorylation of Akt, ERK, GSK-3β and BAD and a significant 6-fold elevation in HO-1 levels. In hypoxic cardiofibroblasts, AN-7 did not activate Akt and ERK beyond the effect of hypoxia alone and induced a limited (~1.5-fold) increase in HO-1. The cell specific differences in kinase activation and in heme oxygenase-1 upregulation may explain, at least in part, the disparate outcome of AN-7 treatment in hypoxic cardiomyocytes and hypoxic cardiofibroblasts.
Collapse
Affiliation(s)
- Vadim Nudelman
- The Felsenstein Medical Research Center, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Muayad A Zahalka
- The Felsenstein Medical Research Center, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Abraham Nudelman
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, Israel.
| | - Ada Rephaeli
- The Felsenstein Medical Research Center, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Gania Kessler-Icekson
- The Felsenstein Medical Research Center, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
8
|
Ameri P, Schiattarella GG, Crotti L, Torchio M, Bertero E, Rodolico D, Forte M, Di Mauro V, Paolillo R, Chimenti C, Torella D, Catalucci D, Sciarretta S, Basso C, Indolfi C, Perrino C. Novel Basic Science Insights to Improve the Management of Heart Failure: Review of the Working Group on Cellular and Molecular Biology of the Heart of the Italian Society of Cardiology. Int J Mol Sci 2020; 21:E1192. [PMID: 32054029 PMCID: PMC7072832 DOI: 10.3390/ijms21041192] [Citation(s) in RCA: 4] [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: 01/02/2020] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Despite important advances in diagnosis and treatment, heart failure (HF) remains a syndrome with substantial morbidity and dismal prognosis. Although implementation and optimization of existing technologies and drugs may lead to better management of HF, new or alternative strategies are desirable. In this regard, basic science is expected to give fundamental inputs, by expanding the knowledge of the pathways underlying HF development and progression, identifying approaches that may improve HF detection and prognostic stratification, and finding novel treatments. Here, we discuss recent basic science insights that encompass major areas of translational research in HF and have high potential clinical impact.
Collapse
Affiliation(s)
- Pietro Ameri
- IRCCS Ospedale Policlinico San Martino—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | | | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, 20149 Milan, Italy;
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Margherita Torchio
- Istituto Auxologico Italiano, IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin, and Laboratory of Cardiovascular Genetics, 20095 Milan, Italy;
| | - Edoardo Bertero
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany;
| | - Daniele Rodolico
- Agostino Gemelli Medical School, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Maurizio Forte
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzili, Italy; (M.F.); (S.S.)
| | - Vittoria Di Mauro
- National Research Council (CNR) Institute of Genetics & Biomedical Research, Milan Unit, 20138 Milan, Italy; (V.D.M.); (D.C.)
- Humanitas Clinical and Research Hospital, 20090 Rozzano (MI), Italy
| | - Roberta Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, 80131 Naples, Italy;
| | - Cristina Chimenti
- Department of Cardiovascular, Respiratory, Nephrologic, and Geriatric Sciences, Sapienza University of Rome, 00100 Rome, Italy;
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Daniele Catalucci
- National Research Council (CNR) Institute of Genetics & Biomedical Research, Milan Unit, 20138 Milan, Italy; (V.D.M.); (D.C.)
- Humanitas Clinical and Research Hospital, 20090 Rozzano (MI), Italy
| | - Sebastiano Sciarretta
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzili, Italy; (M.F.); (S.S.)
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Cristina Basso
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy;
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences & Center of Cardiovascular Research, Magna Graecia University, 88100 Catanzaro, Italy;
- URT-CNR, Magna Graecia University, 88100 Catanzaro, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, 80131 Naples, Italy;
| |
Collapse
|
9
|
Russo M, Guida F, Paparo L, Trinchese G, Aitoro R, Avagliano C, Fiordelisi A, Napolitano F, Mercurio V, Sala V, Li M, Sorriento D, Ciccarelli M, Ghigo A, Hirsch E, Bianco R, Iaccarino G, Abete P, Bonaduce D, Calignano A, Berni Canani R, Tocchetti CG. The novel butyrate derivative phenylalanine-butyramide protects from doxorubicin-induced cardiotoxicity. Eur J Heart Fail 2019; 21:519-528. [PMID: 30843309 DOI: 10.1002/ejhf.1439] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/24/2018] [Accepted: 01/11/2019] [Indexed: 11/10/2022] Open
Abstract
AIMS Butyric acid (BUT), a short chain fatty acid produced daily by the gut microbiota, has proven beneficial in models of cardiovascular diseases. With advancements in cancer survival, an increasing number of patients are at risk of anticancer drug cardiotoxicity. Here we assess whether the novel BUT derivative phenylalanine-butyramide (FBA) protects from doxorubicin (DOXO) cardiotoxicity, by decreasing oxidative stress and improving mitochondrial function. METHODS AND RESULTS In C57BL6 mice, DOXO produced left ventricular dilatation assessed by echocardiography. FBA prevented left ventricular dilatation, fibrosis and cardiomyocyte apoptosis when co-administered with DOXO. DOXO increased atrial natriuretic peptide, brain natriuretic peptide, connective tissue growth factor, and matrix metalloproteinase-2 mRNAs, which were not elevated on co-treatment with FBA. DOXO, but not FBA + DOXO mice, also showed higher nitrotyrosine levels, and increased inducible nitric oxide synthase expression. Accordingly, DOXO hearts showed lower levels of intracellular catalase vs. sham, while pre-treatment with FBA prevented this decrease. We then assessed for reactive oxygen species (ROS) emission: DOXO induced increased activity of mitochondrial superoxide dismutase and higher production of H2 O2 , which were blunted by FBA pre-treatment. FBA also ameliorated mitochondrial state 3 and state 4 respiration rates that were compromised by DOXO. Furthermore, in DOXO animals, the mitochondrial degree of coupling was significantly increased vs. sham, while FBA was able to prevent such increase, contributing to limit ROS production, Finally, FBA reduced DOXO damage in human cellular models, and increased the tumour-killing action of DOXO. CONCLUSIONS Phenylalanine-butyramide protects against experimental doxorubicin cardiotoxicity. Such protection is accompanied by reduction in oxidative stress and amelioration of mitochondrial function.
Collapse
Affiliation(s)
- Michele Russo
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Fiorentina Guida
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy
| | | | - Rosita Aitoro
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy
| | | | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, 'Federico II' University, Naples, Italy
| | - Fabiana Napolitano
- Department of Clinical Medicine and Surgery, 'Federico II' University, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy
| | - Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Mingchuan Li
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, 'Federico II' University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, 'Federico II' University, Naples, Italy.,Interdipartimental Center for Clinical and Translational Research (CIRCET), 'Federico II' University, Naples, Italy
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, 'Federico II' University, Naples, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy
| | - Domenico Bonaduce
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy.,Task Force for the Microbiome Studies, 'Federico II' University, Naples, Italy
| | | | - Roberto Berni Canani
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy.,Task Force for the Microbiome Studies, 'Federico II' University, Naples, Italy.,CEINGE Advanced Biotechnologies, 'Federico II' University, Naples, Italy.,European Laboratory for the Investigation of Food Induced Diseases (ELFID), 'Federico II' University, Naples, Italy
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, 'Federico II' University, Naples, Italy.,Interdipartimental Center for Clinical and Translational Research (CIRCET), 'Federico II' University, Naples, Italy.,Task Force for the Microbiome Studies, 'Federico II' University, Naples, Italy
| |
Collapse
|
10
|
Yusoff SI, Roman M, Lai FY, Eagle-Hemming B, Murphy GJ, Kumar T, Wozniak M. Systematic review and meta-analysis of experimental studies evaluating the organ protective effects of histone deacetylase inhibitors. Transl Res 2019; 205:1-16. [PMID: 30528323 PMCID: PMC6386580 DOI: 10.1016/j.trsl.2018.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 01/07/2023]
Abstract
The clinical efficacy of organ protection interventions are limited by the redundancy of cellular activation mechanisms. Interventions that target epigenetic mechanisms overcome this by eliciting genome wide changes in transcription and signaling. We aimed to review preclinical studies evaluating the organ protection effects of histone deacetylase inhibitors (HDACi) with a view to informing the design of early phase clinical trials. A systematic literature search was performed. Methodological quality was assessed against prespecified criteria. The primary outcome was mortality, with secondary outcomes assessing mechanisms. Prespecified analyses evaluated the effects of likely moderators on heterogeneity. The analysis included 101 experimental studies in rodents (n = 92) and swine (n = 9), exposed to diverse injuries, including: ischemia (n = 72), infection (n = 7), and trauma (n = 22). There were a total of 448 comparisons due to the evaluation of multiple independent interventions within single studies. Sodium valproate (VPA) was the most commonly evaluated HDACi (50 studies, 203 comparisons). All of the studies were judged to have significant methodological limitations. HDACi reduced mortality in experimental models of organ injury (risk ratio = 0.52, 95% confidence interval 0.40-0.68, p < 0.001) without heterogeneity. HDACi administration resulted in myocardial, brain and kidney protection across diverse species and injuries that was attributable to increases in prosurvival cell signaling, and reductions in inflammation and programmed cell death. Heterogeneity in the analyses of secondary outcomes was explained by differences in species, type of injury, HDACi class (Class I better), drug (trichostatin better), and time of administration (at least 6 hours prior to injury better). These findings highlight a potential novel application for HDACi in clinical settings characterized by acute organ injury.
Collapse
Affiliation(s)
- Syabira I Yusoff
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK.
| | - Marius Roman
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Florence Y Lai
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Bryony Eagle-Hemming
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Gavin J Murphy
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Tracy Kumar
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Marcin Wozniak
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| |
Collapse
|
11
|
Duan L, Liu C, Hu J, Liu Y, Wang J, Chen G, Li Z, Chen H. Epigenetic mechanisms in coronary artery disease: The current state and prospects. Trends Cardiovasc Med 2017; 28:311-319. [PMID: 29366539 DOI: 10.1016/j.tcm.2017.12.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/18/2017] [Accepted: 12/23/2017] [Indexed: 12/12/2022]
Abstract
Coronary artery disease (CAD) is the leading cause of morbidity and mortality. CAD has both genetic and environmental causes. In the past two decades, the understanding of epigenetics has advanced swiftly and vigorously. It has been demonstrated that epigenetic modifications are associated with the onset and progression of CAD. This review aims to improve the understanding of the epigenetic mechanisms closely related to CAD and to provide a novel perspective on the onset and development of CAD. Epigenetic changes include DNA methylation, histone modification, microRNA and lncRNA, which are interrelated with critical genes and influence the expression of those genes. In addition, miRNA plays a diverse role in the pathological process of CAD. Numerous studies have found that some cardiac-specific miRNAs have potential as certain diagnostic biomarkers and treatment targets for CAD. In this review, the aberrant epigenetic mechanisms that contribute to CAD will be discussed. We will also provide novel insight into the epigenetic mechanisms that target CAD.
Collapse
Affiliation(s)
- Lian Duan
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Chao Liu
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Junyuan Hu
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Yongmei Liu
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China
| | - Jie Wang
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China.
| | - Guang Chen
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Zhaoling Li
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China
| | - Hengwen Chen
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China
| |
Collapse
|
12
|
Rephaeli A, Tarasenko N, Fibach E, Rozic G, Lubin I, Lipovetsky J, Furman S, Malik Z, Nudelman A. Bi-functional prodrugs of 5-aminolevulinic acid and butyric acid increase erythropoiesis in anemic mice in an erythropoietin-independent manner. Eur J Pharm Sci 2016; 91:91-7. [DOI: 10.1016/j.ejps.2016.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/09/2016] [Accepted: 06/04/2016] [Indexed: 11/28/2022]
|
13
|
Moos WH, Maneta E, Pinkert CA, Irwin MH, Hoffman ME, Faller DV, Steliou K. Epigenetic Treatment of Neuropsychiatric Disorders: Autism and Schizophrenia. Drug Dev Res 2016; 77:53-72. [PMID: 26899191 DOI: 10.1002/ddr.21295] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neuropsychiatric disorders are a heterogeneous group of conditions that often share underlying mitochondrial dysfunction and biological pathways implicated in their pathogenesis, progression, and treatment. To date, these disorders have proven notoriously resistant to molecular-targeted therapies, and clinical options are relegated to interventional types, which do not address the core symptoms of the disease. In this review, we discuss emerging epigenetic-driven approaches using novel acylcarnitine esters (carnitinoids) that act on master regulators of antioxidant and cytoprotective genes and mitophagic pathways. These carnitinoids are actively transported, mitochondria-localizing, biomimetic coenzyme A surrogates of short-chain fatty acids, which inhibit histone deacetylase and may reinvigorate synaptic plasticity and protect against neuronal damage. We outline these neuroprotective effects in the context of treatment of neuropsychiatric disorders such as autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.,SRI Biosciences, A Division of SRI International, Menlo Park, CA, USA
| | - Eleni Maneta
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Carl A Pinkert
- Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, AL, USA.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Michael H Irwin
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Michelle E Hoffman
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Douglas V Faller
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA.,PhenoMatriX, Inc., Boston, MA, USA
| |
Collapse
|
14
|
Abstract
BACKGROUND Evidence reveals that histone deacetylase (HDAC) inhibition has potential for the treatment of inflammatory diseases. The protective effect of HDAC inhibition involves multiple mechanisms. Heme oxygenase-1 (HO-1) is protective in lung injury as a key regulator of antioxidant response. The authors examined whether HDAC inhibition provided protection against ischemia-reperfusion (I/R) lung injury in rats by up-regulating HO-1 activity. METHODS Acute lung injury was induced by producing 40 min of ischemia followed by 60 min of reperfusion in isolated perfused rat lungs. The rats were randomly allotted to control group, I/R group, or I/R + valproic acid (VPA) group with or without an HO-1 activity inhibitor (zinc protoporphyrin IX) (n = 6 per group). RESULTS I/R caused significant increases in the lung edema, pulmonary arterial pressure, lung injury scores, tumor necrosis factor-α, and cytokine-induced neutrophil chemoattractant-1 concentrations in bronchoalveolar lavage fluid. Malondialdehyde levels, carbonyl contents, and myeloperoxidase-positive cells in lung tissue were also significantly increased. I/R stimulated the degradation of inhibitor of nuclear factor-κB-α, nuclear translocation of nuclear factor-κB, and up-regulation of HO-1 activity. Furthermore, I/R decreased B-cell lymphoma-2, heat shock protein 70, acetylated histone H3 protein expression, and increased the caspase-3 activity in the rat lungs. In contrast, VPA treatment significantly attenuated all the parameters of lung injury, oxidative stress, apoptosis, and inflammation. In addition, VPA treatment also enhanced HO-1 activity. Treatment with zinc protoporphyrin IX blocked the protective effect of VPA. CONCLUSIONS VPA protected against I/R-induced lung injury. The protective mechanism may be partly due to enhanced HO-1 activity following HDAC inhibition.
Collapse
|
15
|
el Bahhaj F, Dekker FJ, Martinet N, Bertrand P. Delivery of epidrugs. Drug Discov Today 2014; 19:1337-52. [DOI: 10.1016/j.drudis.2014.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/28/2014] [Accepted: 03/19/2014] [Indexed: 12/22/2022]
|
16
|
Legouis D, Hertig A. La marque des histones dans l’insuffisance rénale aiguë. Nephrol Ther 2013; 9:518-20. [DOI: 10.1016/j.nephro.2013.07.367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 07/21/2013] [Indexed: 11/29/2022]
|
17
|
Steliou K, Boosalis MS, Perrine SP, Sangerman J, Faller DV. Butyrate histone deacetylase inhibitors. Biores Open Access 2013; 1:192-8. [PMID: 23514803 PMCID: PMC3559235 DOI: 10.1089/biores.2012.0223] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In addition to being a part of the metabolic fatty acid fuel cycle, butyrate is also capable of inducing growth arrest in a variety of normal cell types and senescence-like phenotypes in gynecological cancer cells, inhibiting DNA synthesis and cell growth in colonic tumor cell lines, suppressing hTERT mRNA expression and telomerase activity in human prostate cancer cells, and inducing stem cell differentiation and apoptosis by DNA fragmentation. It regulates gene expression by inhibiting histone deacetylases (HDACs), enhances memory recovery and formation in mice, stimulates neurogenesis in the ischemic brain, promotes osteoblast formation, selectively blocks cell replication in transformed cells (compared to healthy cells), and can prevent and treat diet-induced obesity and insulin resistance in mouse models of obesity, as well as stimulate fetal hemoglobin expression in individuals with hematologic diseases such as the thalassemias and sickle-cell disease, in addition to a multitude of other biochemical effects in vivo. However, efforts to exploit the potential of butyrate in the clinical treatment of cancer and other medical disorders are thwarted by its poor pharmacological properties (short half-life and first-pass hepatic clearance) and the multigram doses needed to achieve therapeutic concentrations in vivo. Herein, we review some of the methods used to overcome these difficulties with an emphasis on HDAC inhibition.
Collapse
Affiliation(s)
- Kosta Steliou
- PhenoMatriX, Inc. , Boston, Massachusetts. ; Cancer Research Center, Boston University School of Medicine , Boston, Massachusetts
| | | | | | | | | |
Collapse
|