1
|
Landa MS, Schuman ML, Aisicovich M, Peres Diaz LS, Gironacci MM, García SI, Pirola CJ. Valproate decreases transgenerationally blood pressure by affecting thyrotropin-releasing hormone promoter DNA methylation and gene expression in spontaneously hypertensive rat. Mol Cell Biochem 2025; 480:937-949. [PMID: 38630362 DOI: 10.1007/s11010-024-05001-4] [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: 06/14/2023] [Accepted: 03/25/2024] [Indexed: 02/19/2025]
Abstract
Central TRH, a neuropeptide, is involved in cardiovascular regulation. We demonstrated that the overexpression of diencephalic TRH (dTRH) in SHR rats can be prevented by antisense treatment, normalizing blood pressure (BP). Valproate (VPA) is an inhibitor of histone deacetylases (HDAC) which modulates gene expression through epigenetic modifications such as DNA methylation. AIMS Study the role of HDAC inhibition in the regulation of dTRH gene expression and its effect on the pathogenesis of hypertension. MAIN METHODS We treated 7-weeks-old male and female SHR and WKY rats with VPA for 10 weeks and evaluated BP, dTRH mRNA and methylation gene status. KEY FINDINGS VPA attenuated the elevated BP and dTRH mRNA expression characteristic of SHR. Indeed, we found a significant 62% reduction in dTRH mRNA expression in the SHR + VPA group compared to control SHR. The decrease TRH mRNA expression induced by VPA was confirmed "in vitro" in a primary neuron culture using trichostatin A. With methylation specific PCR we demonstrated a significant increase in TRH promoter DNA methylation level in SHR + VPA group compared to control SHR. After 2 weeks of treatment interruption, rats were mated. Although they did not receive any treatment, the offspring born from VPA-treated SHR parents showed similar changes in BP, dTRH expression and methylation status, implying a transgenerational inheritance. Our findings suggest that dTRH modulation by epigenetics mechanism affects BP and could be inherited by the next generation in SHR rats.
Collapse
Affiliation(s)
- María S Landa
- School of Medicine, Institute of Medical Research A. Lanari, Universidad de Buenos Aires, Combatientes de Malvinas 3150, 1427, Ciudad Autonoma de Buenos Aires, Argentina
- Department of Molecular Cardiology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autonoma de Buenos Aires, Argentina
| | - Mariano L Schuman
- School of Medicine, Institute of Medical Research A. Lanari, Universidad de Buenos Aires, Combatientes de Malvinas 3150, 1427, Ciudad Autonoma de Buenos Aires, Argentina
- Department of Molecular Cardiology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autonoma de Buenos Aires, Argentina
| | - Maia Aisicovich
- School of Medicine, Institute of Medical Research A. Lanari, Universidad de Buenos Aires, Combatientes de Malvinas 3150, 1427, Ciudad Autonoma de Buenos Aires, Argentina
- Department of Molecular Cardiology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autonoma de Buenos Aires, Argentina
| | - Ludmila S Peres Diaz
- School of Medicine, Institute of Medical Research A. Lanari, Universidad de Buenos Aires, Combatientes de Malvinas 3150, 1427, Ciudad Autonoma de Buenos Aires, Argentina
- Department of Molecular Cardiology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autonoma de Buenos Aires, Argentina
| | - Mariela M Gironacci
- Department of Biological Chemistry, Faculty of Pharmacy and Biochemistry, School of Pharmacy and Biochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autonoma de Buenos Aires, Argentina
| | - Silvia I García
- School of Medicine, Institute of Medical Research A. Lanari, Universidad de Buenos Aires, Combatientes de Malvinas 3150, 1427, Ciudad Autonoma de Buenos Aires, Argentina.
- Department of Molecular Cardiology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autonoma de Buenos Aires, Argentina.
- Laboratory of Experimental Medicine, Hospital Alemán, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Carlos J Pirola
- Department of Molecular Genetics and Biology of Complex Diseases, Center for Traslational Research in Health, Maimonides University, Hidalgo 775, 1405, Ciudad Autonoma de Buenos Aires, Argentina.
- National Scientific and Technical Research Council (CONICET), Ciudad Autonoma de Buenos Aires, Argentina.
| |
Collapse
|
2
|
Li Y, Song X, Dai L, Wang Y, Luo Q, Lei L, Pu Y. Mechanism of action of exercise regulating intestinal microflora to improve spontaneous hypertension in rats. BIOMOLECULES & BIOMEDICINE 2025; 25:648-662. [PMID: 39484785 PMCID: PMC12010982 DOI: 10.17305/bb.2024.11174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024]
Abstract
Hypertension is a prevalent cardiovascular disease. Exercise is widely recognized as an effective treatment for hypertension, and it may also influence the composition of the intestinal microflora. However, it remains unclear whether exercise can specifically regulate the intestinal microflora in the context of hypertension treatment. In this study, tail blood pressure in spontaneously hypertensive rats (SHR) was measured using a blood pressure meter after exercise intervention and fecal bacteria transplantation following exercise. Blood lipid levels were assessed using an automatic biochemical analyzer, and 16S rRNA sequencing was employed to analyze the intestinal microflora. Histological examinations of ileal tissue were conducted using HE and Masson staining. Intestinal permeability, inflammatory status, and sympathetic activity were evaluated by measuring the levels of diamine oxidase, D-lactic acid, C-reactive protein, interleukin-6, tumor necrosis factor-α, lipopolysaccharide, norepinephrine, angiotensin II, cyclic adenosine monophosphate, and cyclic guanosine monophosphate. Exercise was found to reduce blood pressure and blood lipid levels in SHR. It also improved the composition of the intestinal microflora, as evidenced by a reduced Firmicutes/Bacteroidetes ratio, an increase in bacteria that produce acetic and butyric acid, and higher Chao 1 and Shannon diversity indices. Furthermore, exercise reduced the thickness of the fibrotic and muscular layers in the ileum, increased the goblet cell/villus ratio and villus length, and decreased intestinal permeability, inflammatory markers, and sympathetic nerve activity. The intestinal microbial flora regulated by exercise demonstrated similar effects on hypertension. In conclusion, exercise appears to regulate the intestinal microflora, and this exercise-induced change in flora may contribute to improvements in hypertension in rats.
Collapse
Affiliation(s)
- Yu Li
- Department of Cardiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xiaoju Song
- Department of Cardiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Lianjing Dai
- Department of Cardiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yangyi Wang
- Department of Cardiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Qiong Luo
- Department of Cardiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Lei Lei
- Department of Cardiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yunfei Pu
- Department of Cardiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
3
|
Brosolo G, Da Porto A, Marcante S, Capilupi F, Bertin N, Vivarelli C, Bulfone L, Vacca A, Catena C, Sechi LA. The role for ω-3 polyunsaturated and short chain fatty acids in hypertension: An updated view on the interaction with gut microbiota. Eur J Pharmacol 2024; 985:177107. [PMID: 39515560 DOI: 10.1016/j.ejphar.2024.177107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
As of 2024, arterial hypertension is still considered the leading modifiable cardiovascular risk factor and, due to high rates of undertreatment and poor blood pressure control, the major contributor to human morbidity and mortality. Development of new treatment options and better interventions in lifestyle correction have become a priority of experimental and clinical research. In the last decades, dietary supplementation of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) and generation of gut microbiota-derived short chain fatty acids (SCFAs) have surged as potential and promising interventions for hypertension and cardiovascular prevention. ω-3 PUFAs are considered "essential" fatty acids that can be obtained only from dietary sources. Although previous intervention trials were not consistent in reporting a significant benefit of ω-3 PUFAs, the recent REDUCE-IT trial has provided robust evidence in support of their role in cardiovascular prevention. Recent studies have also identified the intestinal microbiota as a potential player in the pathophysiology and progression of hypertension. Although this might occur through many pathways, generation of SCFAs that is highly dependent on dietary fiber intake is primarily involved, providing an additional target for the development of novel therapeutic strategies. For these reasons, some scientific societies currently recommend dietary supplementation of ω-3 PUFAs and fiber-containing foods in patients with hypertension. In this narrative review, we summarize the results of studies that examined the effects of ω-3 PUFAs and SCFAs on blood pressure, highlighting the mechanisms of action on the vascular system and their possible impact on hypertension, hypertension-related organ damage and, ultimately, cardiovascular outcomes.
Collapse
Affiliation(s)
- Gabriele Brosolo
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Andrea Da Porto
- Department of Medicine, University of Udine, 33100, Udine, Italy; Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Stefano Marcante
- Department of Medicine, University of Udine, 33100, Udine, Italy.
| | - Filippo Capilupi
- Department of Medicine, University of Udine, 33100, Udine, Italy.
| | - Nicole Bertin
- Department of Medicine, University of Udine, 33100, Udine, Italy; Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Cinzia Vivarelli
- Department of Medicine, University of Udine, 33100, Udine, Italy.
| | - Luca Bulfone
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Antonio Vacca
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Cristiana Catena
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Leonardo A Sechi
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy; Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100, Udine, Italy; Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100, Udine, Italy.
| |
Collapse
|
4
|
Zhang X, Wang Y, Li H, Wang DW, Chen C. Insights into the post-translational modifications in heart failure. Ageing Res Rev 2024; 100:102467. [PMID: 39187021 DOI: 10.1016/j.arr.2024.102467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Heart failure (HF), as the terminal manifestation of multiple cardiovascular diseases, causes a huge socioeconomic burden worldwide. Despite the advances in drugs and medical-assisted devices, the prognosis of HF remains poor. HF is well-accepted as a myriad of subcellular dys-synchrony related to detrimental structural and functional remodelling of cardiac components, including cardiomyocytes, fibroblasts, endothelial cells and macrophages. Through the covalent chemical process, post-translational modifications (PTMs) can coordinate protein functions, such as re-localizing cellular proteins, marking proteins for degradation, inducing interactions with other proteins and tuning enzyme activities, to participate in the progress of HF. Phosphorylation, acetylation, and ubiquitination predominate in the currently reported PTMs. In addition, advanced HF is commonly accompanied by metabolic remodelling including enhanced glycolysis. Thus, glycosylation induced by disturbed energy supply is also important. In this review, firstly, we addressed the main types of HF. Then, considering that PTMs are associated with subcellular locations, we summarized the leading regulation mechanisms in organelles of distinctive cell types of different types of HF, respectively. Subsequently, we outlined the aforementioned four PTMs of key proteins and signaling sites in HF. Finally, we discussed the perspectives of PTMs for potential therapeutic targets in HF.
Collapse
Affiliation(s)
- Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yan Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
5
|
Wang M, Zheng L, Meng Y, Ma S, Zhao D, Xu Y. Broadening horizons: intestinal microbiota as a novel biomarker and potential treatment for hypertensive disorders of pregnancy. Front Cell Infect Microbiol 2024; 14:1446580. [PMID: 39239636 PMCID: PMC11374776 DOI: 10.3389/fcimb.2024.1446580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/24/2024] [Indexed: 09/07/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are severe complications of pregnancy with high morbidity and are a major cause of increased maternal and infant morbidity and mortality. Currently, there is a lack of effective early diagnostic indicators and safe and effective preventive strategies for HDP in clinical practice, except for monitoring maternal blood pressure levels, the degree of proteinuria, organ involvement and fetal conditions. The intestinal microbiota consists of the gut flora and intestinal environment, which is the largest microecosystem of the human body and participates in material and energy metabolism, gene expression regulation, immunity regulation, and other functions. During pregnancy, due to changes in hormone levels and altered immune function, the intestinal microecological balance is affected, triggering HDP. A dysregulated intestinal microenvironment influences the composition and distribution of the gut flora and changes the intestinal barrier, driving beneficial or harmful bacterial metabolites and inflammatory responses to participate in the development of HDP and promote its malignant development. When the gut flora is dysbiotic and affects blood pressure, supplementation with probiotics and dietary fiber can be used to intervene. In this review, the interaction between the intestinal microbiota and HDP was investigated to explore the feasibility of the gut flora as a novel biomarker of HDP and to provide a new strategy and basis for the prevention and treatment of clinical HDP.
Collapse
Affiliation(s)
- Min Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yang Meng
- Jilin Province Product Quality Supervision and Inspection Institute, Changchun, China
| | - Shuai Ma
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Donghai Zhao
- Department of Pathology, Jilin Medical College, Jilin, China
| | - Ying Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Zhou C, Zhao D, Wu C, Wu Z, Zhang W, Chen S, Zhao X, Wu S. Role of histone deacetylase inhibitors in non-neoplastic diseases. Heliyon 2024; 10:e33997. [PMID: 39071622 PMCID: PMC11283006 DOI: 10.1016/j.heliyon.2024.e33997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Background Epigenetic dysregulation has been implicated in the development and progression of a variety of human diseases, but epigenetic changes are reversible, and epigenetic enzymes and regulatory proteins can be targeted using small molecules. Histone deacetylase inhibitors (HDACis), as a class of epigenetic drugs, are widely used to treat various cancers and other diseases involving abnormal gene expression. Results Specially, HDACis have emerged as a promising strategy to enhance the therapeutic effect of non-neoplastic conditions, including neurological disorders, cardiovascular diseases, renal diseases, autoimmune diseases, inflammatory diseases, infectious diseases and rare diseases, along with their related mechanisms. However, their clinical efficacy has been limited by drug resistance and toxicity. Conclusions To date, most clinical trials of HDAC inhibitors have been related to the treatment of cancer rather than the treatment of non-cancer diseases, for which experimental studies are gradually underway. Discussions regarding non-neoplastic diseases often concentrate on specific disease types. Therefore, this review highlights the development of HDACis and their potential therapeutic applications in non-neoplastic diseases, either as monotherapy or in combination with other drugs or therapies.
Collapse
Affiliation(s)
- Chunxiao Zhou
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Dengke Zhao
- Harbin Medical University, Harbin, 150000, China
| | - Chunyan Wu
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Zhimin Wu
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Wen Zhang
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Shilv Chen
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Xindong Zhao
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Shaoling Wu
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| |
Collapse
|
7
|
Mikšiūnas R, Labeit S, Bironaite D. Class I and II Histone Deacetylase Inhibitors as Therapeutic Modulators of Dilated Cardiac Tissue-Derived Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2024; 25:6758. [PMID: 38928463 PMCID: PMC11203858 DOI: 10.3390/ijms25126758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of dilated cardiomyopathy (DCM) is increasing globally, highlighting the need for innovative therapeutic approaches to prevent its onset. In this study, we examined the energetic and epigenetic distinctions between dilated and non-dilated human myocardium-derived mesenchymal stem/stromal cells (hmMSCs) and assessed the effects of class I and II HDAC inhibitors (HDACi) on these cells and their cardiomyogenic differentiation. Cells were isolated from myocardium biopsies using explant outgrowth methods. Mitochondrial and histone deacetylase activities, ATP levels, cardiac transcription factors, and structural proteins were assessed using flow cytometry, PCR, chemiluminescence, Western blotting, and immunohistochemistry. The data suggest that the tested HDAC inhibitors improved acetylation and enhanced the energetic status of both types of cells, with significant effects observed in dilated myocardium-derived hmMSCs. Additionally, the HDAC inhibitors activated the cardiac transcription factors Nkx2-5, HOPX, GATA4, and Mef2C, and upregulated structural proteins such as cardiac troponin T and alpha cardiac actin at both the protein and gene levels. In conclusion, our findings suggest that HDACi may serve as potential modulators of the energetic status and cardiomyogenic differentiation of human heart hmMSCs. This avenue of exploration could broaden the search for novel therapeutic interventions for dilated cardiomyopathy, ultimately leading to improvements in heart function.
Collapse
Affiliation(s)
- Rokas Mikšiūnas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08406 Vilnius, Lithuania;
| | | | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08406 Vilnius, Lithuania;
| |
Collapse
|
8
|
Swiatlowska P, Iskratsch T. Cardiovascular Mechano-Epigenetics: Force-Dependent Regulation of Histone Modifications and Gene Regulation. Cardiovasc Drugs Ther 2024; 38:215-222. [PMID: 36653625 PMCID: PMC10959834 DOI: 10.1007/s10557-022-07422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2022] [Indexed: 01/19/2023]
Abstract
The local mechanical microenvironment impacts on the cell behavior. In the cardiovascular system, cells in both the heart and the vessels are exposed to continuous blood flow, blood pressure, stretching forces, and changing extracellular matrix stiffness. The force-induced signals travel all the way to the nucleus regulating epigenetic changes such as chromatin dynamics and gene expression. Mechanical cues are needed at the very early stage for a faultless embryological development, while later in life, aberrant mechanical signaling can lead to a range of pathologies, including diverse cardiovascular diseases. Hence, an investigation of force-generated epigenetic alteration at different time scales is needed to understand fully the phenotypic changes in disease onset and progression. That being so, cardiovascular mechano-epigenetics emerges as an attractive field of study. Given the rapid advances in this emergent field of research, this short review aims to provide an analysis of the state of knowledge of force-induced epigenetic changes in the cardiovascular field.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK.
| |
Collapse
|
9
|
Ray A, Stelloh C, Liu Y, Meyer A, Geurts AM, Cowley A, Greene AS, Liang M, Rao S. Histone Modifications and Their Contributions to Hypertension. Hypertension 2024; 81:229-239. [PMID: 38031837 PMCID: PMC11229175 DOI: 10.1161/hypertensionaha.123.21755] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Essential hypertension, a multifaceted disorder, is a worldwide health problem. A complex network of genetic, epigenetic, physiological, and environmental components regulates blood pressure (BP), and any dysregulation of this network may result in hypertension. Growing evidence suggests a role for epigenetic factors in BP regulation. Any alterations in the expression or functions of these epigenetic regulators may dysregulate various determinants of BP, thereby promoting the development of hypertension. Histone posttranslational modifications are critical epigenetic regulators that have been implicated in hypertension. Several studies have demonstrated a clear association between the increased expression of some histone-modifying enzymes, especially HDACs (histone deacetylases), and hypertension. In addition, treatment with HDAC inhibitors lowers BP in hypertensive animal models, providing an excellent opportunity to design new drugs to treat hypertension. In this review, we discuss the potential contribution of different histone modifications to the regulation of BP.
Collapse
Affiliation(s)
- Atrayee Ray
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Cary Stelloh
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
| | - Yong Liu
- Department of Physiology, University of Arizona, Tucson, AZ 85721
| | - Alison Meyer
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
| | - Aron M Geurts
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Allen Cowley
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Mingyu Liang
- Department of Physiology, University of Arizona, Tucson, AZ 85721
| | - Sridhar Rao
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
- Department of Pediatrics, Section of Hematology/Oncology/Transplantation, Medical College of Wisconsin, WI 53226, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, WI, 53226, USA
| |
Collapse
|
10
|
Cai M, Lin L, Jiang F, Peng Y, Li S, Chen L, Lin Y. Gut microbiota changes in patients with hypertension: A systematic review and meta-analysis. J Clin Hypertens (Greenwich) 2023; 25:1053-1068. [PMID: 37853925 PMCID: PMC10710550 DOI: 10.1111/jch.14722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 10/20/2023]
Abstract
Hypertension is a major public health issue worldwide. The imbalance of gut microbiota is thought to play an important role in the pathogenesis of hypertension. The authors conducted the systematic review and meta-analysis to clarify the relationship between gut microbiota and hypertension through conducting an electronic search in six databases. Our meta-analysis included 19 studies and the results showed that compared with healthy controls, Shannon significantly decreased in hypertension [SMD = -0.13, 95%CI (-0.22, -0.04), p = .007]; however, Simpson [SMD = -0.01, 95%CI (-0.14, 0.12), p = .87], ACE [SMD = 0.18, 95%CI (-0.06, 0.43), p = .14], and Chao1 [SMD = 0.11, 95%CI (-0.01, 0.23), p = .08] did not differ significantly between hypertension and healthy controls. The F/B ratio significantly increased in hypertension [SMD = 0.84, 95%CI (0.10, 1.58), p = .03]. In addition, Shannon index was negatively correlated with hypertension [r = -0.12, 95%CI (-0.19, -0.05)], but had no significant correlation with SBP [r = 0.10, 95%CI (-0.19, 0.37)] and DBP [r = -0.39, 95%CI (-0.73, 0.12)]. At the phylum level, the relative abundance of Firmicutes [SMD = -0.01, 95%CI (-0.37, 0.34), p = .94], Bacteroidetes [SMD = -0.15, 95%CI (-0.44, 0.14), p = .30], Proteobacteria [SMD = 0.25, 95%CI (-0.01, 0.51), p = .06], and Actinobacteria [SMD = 0.21, 95%CI (-0.11, 0.53), p = .21] did not differ significantly between hypertension and healthy controls. At the genus level, compared with healthy controls, the relative abundance of Faecalibacterium decreased significantly [SMD = -0.16, 95%CI (-0.28, -0.04), p = .01], while the Streptococcus [SMD = 0.20, 95%CI (0.08, 0.32), p = .001] and Enterococcus [SMD = 0.20, 95%CI (0.08, 0.33), p = .002] significantly increased in hypertension. Available evidence suggests that hypertensive patients may have an imbalance of gut microbiota. However, it still needs further validation by large sample size studies of high quality.
Collapse
Affiliation(s)
- Meiling Cai
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| | - Lingyu Lin
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Fei Jiang
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| | - Yanchun Peng
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Sailan Li
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Liangwan Chen
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| | - Yanjuan Lin
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| |
Collapse
|
11
|
Habibian JS, Bolino M, Qian A, Woolsey R, Quilici D, Petereit J, Ferguson BS. Class I HDAC inhibitors attenuate dexamethasone-induced muscle atrophy via increased protein kinase C (PKC) delta phosphorylation. Cell Signal 2023; 110:110815. [PMID: 37478958 PMCID: PMC10528066 DOI: 10.1016/j.cellsig.2023.110815] [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: 01/26/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Skeletal muscle atrophy is defined by wasting or decrease in muscle mass owing to injury, aging, malnutrition, chronic disuse, or physical consequences of chronic illness. Under normal physiological conditions, a network of signal transduction pathways serves to balance muscle protein synthesis and proteolysis; however, metabolic shifts occur from protein synthesis to protein degradation that leads to a reduction in cross-sectional myofibers and can result in loss of skeletal muscle mass (atrophy) over time. Recent evidence highlights posttranslational modifications (PTMs) such as acetylation and phosphorylation in contractile dysfunction and muscle wasting. Indeed, histone deacetylase (HDAC) inhibitors have been shown to attenuate muscle atrophy and delay muscle damage in response to nutrient deprivation, in models of metabolic dysfunction and genetic models of muscle disease (e.g., muscle dystrophy). Despite our current understanding of lysine acetylation in muscle physiology, a role for HDACs in the regulation of muscle signal transduction remains a 'black box.' Using C2C12 myotubes stimulated with dexamethasone (Dex) as a model of muscle atrophy, we report that protein kinase C delta (PKCδ) phosphorylation decreased at threonine 505 (T505) and serine 643 (S643) in myotubes in response to muscle atrophy; these residues are important for PKCδ activity. Interestingly, PKCδ phosphorylation was restored/increased in myotubes treated with a pan-HDAC inhibitor or a class I selective HDAC inhibitor targeting HDACs1, -2, and - 3 in response to Dex. Moreover, we observed that Dex induced atrophy in skeletal muscle tissue in mice; this reduction in atrophy occurred rapidly, with weight loss noted by day 3 post-Dex and muscle weight loss noted by day 7. Similar to our findings in C2C12 myotubes, Dex attenuated phosphorylation of PKCδ at S643, while HDAC inhibition restored or increased PKCδ phosphorylation at both T505 and S643 in the tibialis anterior. Consistent with this hypothesis, we report that HDAC inhibition could not restore myotube size in response to Dex in the presence of a PKCδ inhibitor or when overexpressing a dominant negative PKCδ. Additionally, the overexpression of a constitutively active PKCδ prevented Dex-induced myotube atrophy. Combined, these data suggest that HDACs regulate muscle physiology via changes in intracellular signaling, namely PKCδ phosphorylation. Whether HDACs regulate PKCδ through canonical (e.g. gene-mediated regulation of phosphatases) or non-canonical (e.g. direct deacetylation of PKCδ to change phosphorylation states) mechanisms remain unclear and future research is needed to clarify this point.
Collapse
Affiliation(s)
- Justine S Habibian
- Department of Nutrition, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Pharmacology and Physiology, The University of Nevada Reno, Reno, NV 89557, USA
| | - Matthew Bolino
- Department of Nutrition, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV 89557, United States of America
| | - Anthony Qian
- Cellular and Molecular Pharmacology and Physiology, The University of Nevada Reno, Reno, NV 89557, USA
| | - Rebekah Woolsey
- Mick Hitchcock, Ph.D. Nevada Proteomics Center (RRID:SCR_017761), The University of Nevada Reno, Reno, NV 89557, USA
| | - David Quilici
- Mick Hitchcock, Ph.D. Nevada Proteomics Center (RRID:SCR_017761), The University of Nevada Reno, Reno, NV 89557, USA
| | - Juli Petereit
- Nevada Bioinformatics Center (RRID:SCR_017802), The University of Nevada Reno, Reno, NV 89557, USA
| | - Bradley S Ferguson
- Department of Nutrition, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV 89557, United States of America; Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, The University of Nevada Reno, Reno, NV 89557, United States of America.
| |
Collapse
|
12
|
Wołowiec A, Wołowiec Ł, Grześk G, Jaśniak A, Osiak J, Husejko J, Kozakiewicz M. The Role of Selected Epigenetic Pathways in Cardiovascular Diseases as a Potential Therapeutic Target. Int J Mol Sci 2023; 24:13723. [PMID: 37762023 PMCID: PMC10531432 DOI: 10.3390/ijms241813723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Epigenetics is a rapidly developing science that has gained a lot of interest in recent years due to the correlation between characteristic epigenetic marks and cardiovascular diseases (CVDs). Epigenetic modifications contribute to a change in gene expression while maintaining the DNA sequence. The analysis of these modifications provides a thorough insight into the cardiovascular system from its development to its further functioning. Epigenetics is strongly influenced by environmental factors, including known cardiovascular risk factors such as smoking, obesity, and low physical activity. Similarly, conditions affecting the local microenvironment of cells, such as chronic inflammation, worsen the prognosis in cardiovascular diseases and additionally induce further epigenetic modifications leading to the consolidation of unfavorable cardiovascular changes. A deeper understanding of epigenetics may provide an answer to the continuing strong clinical impact of cardiovascular diseases by improving diagnostic capabilities, personalized medical approaches and the development of targeted therapeutic interventions. The aim of the study was to present selected epigenetic pathways, their significance in cardiovascular diseases, and their potential as a therapeutic target in specific medical conditions.
Collapse
Affiliation(s)
- Anna Wołowiec
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Łukasz Wołowiec
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Grzegorz Grześk
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Albert Jaśniak
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Joanna Osiak
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Jakub Husejko
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Mariusz Kozakiewicz
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| |
Collapse
|
13
|
Jin G, Wang K, Zhao Y, Yuan S, He Z, Zhang J. Targeting histone deacetylases for heart diseases. Bioorg Chem 2023; 138:106601. [PMID: 37224740 DOI: 10.1016/j.bioorg.2023.106601] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Histone deacetylases (HDACs) are responsible for the deacetylation of lysine residues in histone or non-histone substrates, leading to the regulation of many biological functions, such as gene transcription, translation and remodeling chromatin. Targeting HDACs for drug development is a promising way for human diseases, including cancers and heart diseases. In particular, numerous HDAC inhibitors have revealed potential clinical value for the treatment of cardiac diseases in recent years. In this review, we systematically summarize the therapeutic roles of HDAC inhibitors with different chemotypes on heart diseases. Additionally, we discuss the opportunities and challenges in developing HDAC inhibitors for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Gang Jin
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China
| | - Kaiyue Wang
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China
| | - Yaohui Zhao
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Zhangxu He
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China.
| | - Jingyu Zhang
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China.
| |
Collapse
|
14
|
Nesci A, Carnuccio C, Ruggieri V, D'Alessandro A, Di Giorgio A, Santoro L, Gasbarrini A, Santoliquido A, Ponziani FR. Gut Microbiota and Cardiovascular Disease: Evidence on the Metabolic and Inflammatory Background of a Complex Relationship. Int J Mol Sci 2023; 24:ijms24109087. [PMID: 37240434 DOI: 10.3390/ijms24109087] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Several studies in recent years have demonstrated that gut microbiota-host interactions play an important role in human health and disease, including inflammatory and cardiovascular diseases. Dysbiosis has been linked to not only well-known inflammatory diseases, such as inflammatory bowel diseases, rheumatoid arthritis, and systemic lupus erythematous, but also to cardiovascular risk factors, such as atherosclerosis, hypertension, heart failure, chronic kidney disease, obesity, and type 2 diabetes mellitus. The ways the microbiota is involved in modulating cardiovascular risk are multiple and not only related to inflammatory mechanisms. Indeed, human and the gut microbiome cooperate as a metabolically active superorganism, and this affects host physiology through metabolic pathways. In turn, congestion of the splanchnic circulation associated with heart failure, edema of the intestinal wall, and altered function and permeability of the intestinal barrier result in the translocation of bacteria and their products into the systemic circulation, further enhancing the pro-inflammatory conditions underlying cardiovascular disorders. The aim of the present review is to describe the complex interplay between gut microbiota, its metabolites, and the development and evolution of cardiovascular diseases. We also discuss the possible interventions intended to modulate the gut microbiota to reduce cardiovascular risk.
Collapse
Affiliation(s)
- Antonio Nesci
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Claudia Carnuccio
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Vittorio Ruggieri
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Alessia D'Alessandro
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Angela Di Giorgio
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Luca Santoro
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center (CEMAD), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Angelo Santoliquido
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Digestive Disease Center (CEMAD), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
15
|
Does the Composition of Gut Microbiota Affect Hypertension? Molecular Mechanisms Involved in Increasing Blood Pressure. Int J Mol Sci 2023; 24:ijms24021377. [PMID: 36674891 PMCID: PMC9863380 DOI: 10.3390/ijms24021377] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Arterial hypertension is a chronic disease which is very prevalent contemporarily. The aim of this review was to investigate the impact of gut microbiota on the development and potential treatment of hypertension, taking into consideration underlying molecular mechanisms. The bacteria present in the intestines have the ability to secrete different metabolites, which might play a significant role in the regulation of blood pressure. The most important include short-chain fatty acids (SCFAs), vasoactive hormones, trimethylamine (TMA) and trimethylamine N-oxide (TMAO) and uremic toxins, such as indoxyl sulfate (IS) and p-cresyl sulfate (PCS). Their action in regulating blood pressure is mainly based on their pro- or anti-inflammatory function. The use of specifically formulated probiotics to modify the composition of gut microbiota might be a beneficial way of supportive treatment of hypertension; however, further research on this topic is needed to choose the species of bacteria that could induce the hypotensive pattern.
Collapse
|
16
|
Shanmukha KD, Paluvai H, Lomada SK, Gokara M, Kalangi SK. Histone deacetylase (HDACs) inhibitors: Clinical applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:119-152. [DOI: 10.1016/bs.pmbts.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
17
|
Li Y, Zhu M, Liu Y, Luo B, Cui J, Huang L, Chen K, Liu Y. The oral microbiota and cardiometabolic health: A comprehensive review and emerging insights. Front Immunol 2022; 13:1010368. [PMID: 36466857 PMCID: PMC9716288 DOI: 10.3389/fimmu.2022.1010368] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/26/2022] [Indexed: 08/26/2023] Open
Abstract
There is mounting evidence demonstrating that oral dysbiosis causes periodontal disease and promotes the development of cardiovascular disease. The advancement of omics techniques has driven the optimization of oral microbiota species analysis and has provided a deeper understanding of oral pathogenic bacteria. A bi-directional relationship exists between the oral microbiota and the host, and oral-gut microbiota transfer is known to alter the composition of the gut microbiota and may cause local metabolic disorders. Furthermore, cardiovascular health can also be highly affected by oral microbiota functions and metabolites, including short-chain fatty acids (SCFAs), nitric oxide (NO), hydrogen sulfide (H2S), and some lipid metabolites. Studies have found that trimethylamine oxide (TMAO) may have adverse effects on cardiovascular health, whereas SCFAs, NO, and H2S have cardioprotective effects. SCFAs and H2S exert varying oral and cardiovascular effects, however reports on this specific topic remain controversial. Previous evidences are accustomed to summarizing the functions of oral microbiota in the context of periodontitis. The direct relationship between oral microbiota and cardiovascular diseases is insufficient. By systematically summarizing the methods associated with oral microbiota transplantation (OMT), this review facilitates an investigation into the causal links between oral microbiota and cardiovascular disease. The concomitant development of omics, bioinformatics, bacterial culture techniques, and microbiota transplantation techniques is required to gain a deeper understanding of the relationship between oral microbiota and cardiovascular disease occurrence.
Collapse
Affiliation(s)
- Yiwen Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Mengmeng Zhu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Liu
- The Second Department of Gerontology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Binyu Luo
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Cui
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- China Center for Evidence-based Medicine of Traditional Chinese Medicine (TCM), China Academy of Chinese Medical Sciences, Beijing, China
| | - Keji Chen
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Sun D, Xiang H, Yan J, He L. Intestinal microbiota: A promising therapeutic target for hypertension. Front Cardiovasc Med 2022; 9:970036. [PMID: 36457803 PMCID: PMC9705378 DOI: 10.3389/fcvm.2022.970036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/28/2022] [Indexed: 10/29/2023] Open
Abstract
Hypertension has developed into an escalating serious global public health problem with multiple and unclear pathophysiological mechanisms. Recent studies have identified intestinal microbiota as a key perpetrator of hypertension through a variety of mechanisms. In this review, we highlight the potential roles of the intestinal microbiota and its metabolites in the development of hypertension, as well as the therapeutic potential for targeting intestinal microbiomes. We also shed light on the main limitations and challenges of the current research and suggest directions for future investigations. Finally, we discuss the development of accurate and personalized preventive and therapeutic strategies for hypotension by the modulation of intestinal microbes and metabolites.
Collapse
Affiliation(s)
- Dating Sun
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Hui Xiang
- Infectious Disease Department, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liqun He
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| |
Collapse
|
19
|
Increasing angiotensin-converting enzyme 1 regulated by histone 3 lysine 27 hyperacetylation in high-fat diet-induced hypertensive rat kidney. J Hypertens 2022; 40:1969-1978. [PMID: 35969203 DOI: 10.1097/hjh.0000000000003210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Obesity is a key risk factor of hypertension. Angiotensin-converting enzyme 1 (ACE1) is a key enzyme involved in the renin-angiotensin-aldosterone system (RAAS), which contributes to obesity-related hypertension (OrHTN). Emerging evidence has shown that histone acetylation is also involved in OrHTN. As kidney is an effector organ that activates the RAAS by secreting renin after hypertension occurs, this study aimed to explore the regulatory role of histone acetylation on renal RAAS expression. METHODS Nineteen male Wistar rats were randomly divided into a control group ( n = 9, fed normal chow) and a high-fat diet (HFD) group ( n = 10, fed HFD for 16 weeks). The renal transcriptome and histone acetylation spectrum was analyzed by RNA sequencing and tandem mass spectrometry and was further confirmed by RT-qPCR, western blot, and immunohistochemistry. Then, chromatin immunoprecipitation (ChIP)-qPCR analysis was performed for the detection of DNA-protein interaction. RESULTS After 16-week HFD, the rats became obese with increased plasma triglyceride and high blood pressure. Increased ACE1 and histone 3 lysine 27 acetylation (H3K27ac) expression levels were found in OrHTN rat kidneys. The following ChIP-qPCR analysis illustrated that the upregulation of ACE1 transcription was mediated by increased H3K27ac. CONCLUSION H3K27ac could be an important histone acetylation site that activates renal ACE1 in HFD-induced hypertensive rats.
Collapse
|
20
|
Elrakaybi A, Laubner K, Zhou Q, Hug MJ, Seufert J. Cardiovascular protection by SGLT2 inhibitors - Do anti-inflammatory mechanisms play a role? Mol Metab 2022; 64:101549. [PMID: 35863639 PMCID: PMC9352970 DOI: 10.1016/j.molmet.2022.101549] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Metabolic syndrome and related metabolic disturbances represent a state of low-grade inflammation, which accelerates insulin resistance, type 2 diabetes (T2D) and cardiovascular disease (CVD) progression. Among antidiabetic medications, sodium glucose co-transporter (SGLT) 2 inhibitors are the only agents which showed remarkable reductions in heart failure (HF) hospitalizations and major cardiovascular endpoints (MACE) as well as renal endpoints regardless of diabetes status in large randomized clinical outcome trials (RCTs). Although the exact mechanisms underlying these benefits are yet to be established, growing evidence suggests that modulating inflammation by SGLT2 inhibitors may play a key role. SCOPE OF REVIEW In this manuscript, we summarize the current knowledge on anti-inflammatory effects of SGLT2 inhibitors as one of the mechanisms potentially mediating their cardiovascular (CV) benefits. We introduce the different metabolic and systemic actions mediated by these agents which could mitigate inflammation, and further present the signalling pathways potentially responsible for their proposed direct anti-inflammatory effects. We also discuss controversies surrounding some of these mechanisms. MAJOR CONCLUSIONS SGLT2 inhibitors are promising anti-inflammatory agents by acting either indirectly via improving metabolism and reducing stress conditions or via direct modulation of inflammatory signalling pathways. These effects were achieved, to a great extent, in a glucose-independent manner which established their clinical use in HF patients with and without diabetes.
Collapse
Affiliation(s)
- Asmaa Elrakaybi
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Department of Clinical Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Katharina Laubner
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Qian Zhou
- Department of Cardiology and Angiology I, Heart Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Department of Cardiology, University Hospital Basel, 4031 Basel, Switzerland
| | - Martin J Hug
- Pharmacy, Medical Centre - University of Freiburg, 79106 Freiburg, Germany
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
21
|
Lakshmanan AP, Murugesan S, Al Khodor S, Terranegra A. The potential impact of a probiotic: Akkermansia muciniphila in the regulation of blood pressure—the current facts and evidence. Lab Invest 2022; 20:430. [PMID: 36153618 PMCID: PMC9509630 DOI: 10.1186/s12967-022-03631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
Abstract
Akkermansia muciniphila (A. muciniphila) is present in the human gut microbiota from infancy and gradually increases in adulthood. The potential impact of the abundance of A. muciniphila has been studied in major cardiovascular diseases including elevated blood pressure or hypertension (HTN). HTN is a major factor in premature death worldwide, and approximately 1.28 billion adults aged 30–79 years have hypertension. A. muciniphila is being considered a next-generation probiotic and though numerous studies had highlighted the positive role of A. muciniphila in lowering/controlling the HTN, however, few studies had highlighted the negative impact of increased abundance of A. muciniphila in the management of HTN. Thus, in the review, we aimed to discuss the current facts, evidence, and controversy about the role of A. muciniphila in the pathophysiology of HTN and its potential effect on HTN management/regulation, which could be beneficial in identifying the drug target for the management of HTN.
Collapse
|
22
|
Han Y, Nie J, Wang DW, Ni L. Mechanism of histone deacetylases in cardiac hypertrophy and its therapeutic inhibitors. Front Cardiovasc Med 2022; 9:931475. [PMID: 35958418 PMCID: PMC9360326 DOI: 10.3389/fcvm.2022.931475] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/06/2022] [Indexed: 12/03/2022] Open
Abstract
Cardiac hypertrophy is a key process in cardiac remodeling development, leading to ventricle enlargement and heart failure. Recently, studies show the complicated relation between cardiac hypertrophy and epigenetic modification. Post-translational modification of histone is an essential part of epigenetic modification, which is relevant to multiple cardiac diseases, especially in cardiac hypertrophy. There is a group of enzymes related in the balance of histone acetylation/deacetylation, which is defined as histone acetyltransferase (HAT) and histone deacetylase (HDAC). In this review, we introduce an important enzyme family HDAC, a key regulator in histone deacetylation. In cardiac hypertrophy HDAC I downregulates the anti-hypertrophy gene expression, including Kruppel-like factor 4 (Klf4) and inositol-5 phosphatase f (Inpp5f), and promote the development of cardiac hypertrophy. On the contrary, HDAC II binds to myocyte-specific enhancer factor 2 (MEF2), inhibit the assemble ability to HAT and protect against cardiac hypertrophy. Under adverse stimuli such as pressure overload and calcineurin stimulation, the HDAC II transfer to cytoplasm, and MEF2 can bind to nuclear factor of activated T cells (NFAT) or GATA binding protein 4 (GATA4), mediating inappropriate gene expression. HDAC III, also known as SIRTs, can interact not only to transcription factors, but also exist interaction mechanisms to other HDACs, such as HDAC IIa. We also present the latest progress of HDAC inhibitors (HDACi), as a potential treatment target in cardiac hypertrophy.
Collapse
Affiliation(s)
- Yu Han
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- *Correspondence: Dao Wen Wang,
| | - Li Ni
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- Li Ni,
| |
Collapse
|
23
|
Sirtuin1 inhibitor attenuates hypertension in spontaneously hypertensive rats: role of Giα proteins and nitroxidative stress. J Hypertens 2022; 40:1314-1326. [PMID: 35762472 DOI: 10.1097/hjh.0000000000003143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We recently showed that vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHR) exhibit overexpression of Sirtuin1 (Sirt1) that contributes to the enhanced expression of Giα proteins implicated in the development of hypertension in SHR. METHOD The present study investigated if the inhibition of Sirt1 could also ameliorate hypertension in SHR and explore the underlying molecular mechanisms. For this study, a selective inhibitor of Sirt1, EX-527 (5 mg/kg of body weight), was injected intraperitoneally into 8-week-old SHR and age-matched Wistar Kyoto (WKY) rats twice per week for 3 weeks. The blood pressure (BP) and heart rate was measured twice a week by the CODA noninvasive tail cuff method. RESULTS The high BP and augmented heart rate in SHR was significantly attenuated by EX-527 treatment, which was associated with the suppression of the overexpression of Sirt1 and Giα proteins in heart, VSMC and aorta. In addition, the enhanced levels of superoxide anion, NADPH oxidase activity, overexpression of NADPH oxidase subunits and FOXO1 were attenuated and the decreased levels of endothelial nitric oxide synthase (eNOS), nitric oxide and increased levels of peroxynitrite (ONOO-) and tyrosine nitration in VSMC from SHR were restored to control levels by EX-527 treatment. Furthermore, knockdown of FOXO1 by siRNA also attenuated the overexpression of Giα-2 and NADPH oxidase subunit proteins and restored the decreased expression of eNOS in VSMC from SHR. CONCLUSION These results suggest that the inhibition of overexpressed Sirt1 and its target FOXO1 through decreasing the enhanced levels of Giα proteins and nitro-oxidative stress attenuates the high BP in SHR.
Collapse
|
24
|
Wong HJ, Lim WH, Ng CH, Tan DJH, Bonney GK, Kow AWC, Huang DQ, Siddiqui MS, Noureddin M, Syn N, Muthiah MD. Predictive and Prognostic Roles of Gut Microbial Variation in Liver Transplant. Front Med (Lausanne) 2022; 9:873523. [PMID: 35620719 PMCID: PMC9127379 DOI: 10.3389/fmed.2022.873523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Patients undergoing liver transplant (LTX) typically confront a challenging postoperative journey. A dysbiotic gut microbiome is associated with the development of complications, including post-LTX allograft rejection, metabolic diseases and de novo or recurrent cancer. A major explanation of this are the bipartite interactions between the gut microbiota and host immunity, which modulates the alloimmune response towards the liver allograft. Furthermore, bacterial translocation from dysbiosis causes pathogenic changes in the concentrations of microbial metabolites like lipopolysaccharides, short-chain fatty acids (SCFAs) and Trimethylamine-N-Oxide, with links to cardiovascular disease development and diabetes mellitus. Gut dysbiosis also disrupts bile acid metabolism, with implications for various post-LTX metabolic diseases. Certain taxonomy of microbiota such as lactobacilli, F.prausnitzii and Bacteroides appear to be associated with these undesired outcomes. As such, an interesting but as yet unproven hypothesis exists as to whether induction of a “beneficial” composition of gut microbiota may improve prognosis in LTX patients. Additionally, there are roles of the microbiome as predictive and prognostic indicators for clinicians in improving patient care. Hence, the gut microbiome represents an exceptionally exciting avenue for developing novel prognostic, predictive and therapeutic applications.
Collapse
Affiliation(s)
- Hon Jen Wong
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore
| | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Glenn K Bonney
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore.,Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, National University Hospital Singapore, Singapore, Singapore
| | - Alfred W C Kow
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore.,Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, National University Hospital Singapore, Singapore, Singapore
| | - Daniel Q Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Mazen Noureddin
- Cedars-Sinai Fatty Liver Program, Division of Digestive and Liver Diseases, Department of Medicine, Comprehensive Transplant Centre, Cedars-Sinai Medical Centre, Los Angeles, CA, United States
| | - Nicholas Syn
- National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Mark D Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| |
Collapse
|
25
|
Zinc-dependent histone deacetylases: Potential therapeutic targets for arterial hypertension. Biochem Pharmacol 2022; 202:115111. [DOI: 10.1016/j.bcp.2022.115111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022]
|
26
|
Kolski-Andreaco A, Balut CM, Bertuccio CA, Wilson AS, Rivers WM, Liu X, Gandley RE, Straub AC, Butterworth MB, Binion D, Devor DC. Histone deacetylase inhibitors (HDACi) increase expression of KCa2.3 (SK3) in primary microvascular endothelial cells. Am J Physiol Cell Physiol 2022; 322:C338-C353. [PMID: 35044858 PMCID: PMC8858676 DOI: 10.1152/ajpcell.00409.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The small conductance calcium-activated potassium channel (KCa2.3) has long been recognized for its role in mediating vasorelaxation through the endothelium-derived hyperpolarization (EDH) response. Histone deacetylases (HDACs) have been implicated as potential modulators of blood pressure and histone deacetylase inhibitors (HDACi) are being explored as therapeutics for hypertension. Herein, we show that HDACi increase KCa2.3 expression when heterologously expressed in HEK cells and endogenously expressed in primary cultures of human umbilical vein endothelial cells (HUVECs) and human intestinal microvascular endothelial cells (HIMECs). When primary endothelial cells were exposed to HDACi, KCa2.3 transcripts, subunits, and functional current are increased. Quantitative RT-PCR (qPCR) demonstrated increased KCa2.3 mRNA following HDACi, confirming transcriptional regulation of KCa2.3 by HDACs. By using pharmacological agents selective for different classes of HDACs, we discriminated between cytoplasmic and epigenetic modulation of KCa2.3. Biochemical analysis revealed an association between the cytoplasmic HDAC6 and KCa2.3 in immunoprecipitation studies. Specifically inhibiting HDAC6 increases expression of KCa2.3. In addition to increasing the expression of KCa2.3, we show that nonspecific inhibition of HDACs causes an increase in the expression of the molecular chaperone Hsp70 in endothelial cells. When Hsp70 is inhibited in the presence of HDACi, the magnitude of the increase in KCa2.3 expression is diminished. Finally, we show a slower rate of endocytosis of KCa2.3 as a result of exposure of primary endothelial cells to HDACi. These data provide the first demonstrated approach to increase KCa2.3 channel number in endothelial cells and may partially account for the mechanism by which HDACi induce vasorelaxation.
Collapse
Affiliation(s)
| | - Corina M. Balut
- 1Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Annette S. Wilson
- 2Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William M. Rivers
- 2Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiaoning Liu
- 1Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robin E. Gandley
- 3Department of Obstetrics and Gynecology and Reproductive Sciences, Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam C. Straub
- 4Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - David Binion
- 2Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel C. Devor
- 1Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
27
|
Huang Q, Cai G, Liu T, Liu Z. Relationships Among Gut Microbiota, Ischemic Stroke and Its Risk Factors: Based on Research Evidence. Int J Gen Med 2022; 15:2003-2023. [PMID: 35795301 PMCID: PMC9252587 DOI: 10.2147/ijgm.s353276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/08/2022] [Indexed: 11/27/2022] Open
Abstract
Stroke is a highly lethal disease and disabling illness while ischemic stroke accounts for the majority of stroke. It has been found that inflammation plays a key role in the initiation and progression of stroke, and atherosclerotic plaque rupture is considered to be the leading cause of ischemic stroke. Furthermore, chronic inflammatory diseases, such as obesity, type 2 diabetes mellitus (T2DM) and hypertension, are also considered as the high-risk factors for stroke. Recently, the topic on how gut microbiota affects human health has aroused great concern. The initiation and progression of ischemic stroke has been found to have close relation with gut microbiota dysbiosis. Hence, this manuscript briefly summarizes the roles of gut microbiota in ischemic stroke and its related risk factors, and the practicability of preventing and alleviating ischemic stroke by reconstructing gut microbiota.
Collapse
Affiliation(s)
- Qinhong Huang
- First Clinical School, Guangzhou Medical University, Guangzhou, 511436, People’s Republic of China
| | - Guannan Cai
- First Clinical School, Guangzhou Medical University, Guangzhou, 511436, People’s Republic of China
| | - Ting Liu
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, People’s Republic of China
- Correspondence: Ting Liu; Zhihua Liu, Email ;
| | - Zhihua Liu
- Department of Anorectal Surgery, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, People’s Republic of China
| |
Collapse
|
28
|
Pozo MR, Meredith GW, Entcheva E. Human iPSC-Cardiomyocytes as an Experimental Model to Study Epigenetic Modifiers of Electrophysiology. Cells 2022; 11:200. [PMID: 35053315 PMCID: PMC8774228 DOI: 10.3390/cells11020200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 02/04/2023] Open
Abstract
The epigenetic landscape and the responses to pharmacological epigenetic regulators in each human are unique. Classes of epigenetic writers and erasers, such as histone acetyltransferases, HATs, and histone deacetylases, HDACs, control DNA acetylation/deacetylation and chromatin accessibility, thus exerting transcriptional control in a tissue- and person-specific manner. Rapid development of novel pharmacological agents in clinical testing-HDAC inhibitors (HDACi)-targets these master regulators as common means of therapeutic intervention in cancer and immune diseases. The action of these epigenetic modulators is much less explored for cardiac tissue, yet all new drugs need to be tested for cardiotoxicity. To advance our understanding of chromatin regulation in the heart, and specifically how modulation of DNA acetylation state may affect functional electrophysiological responses, human-induced pluripotent stem-cell-derived cardiomyocyte (hiPSC-CM) technology can be leveraged as a scalable, high-throughput platform with ability to provide patient-specific insights. This review covers relevant background on the known roles of HATs and HDACs in the heart, the current state of HDACi development, applications, and any adverse cardiac events; it also summarizes relevant differential gene expression data for the adult human heart vs. hiPSC-CMs along with initial transcriptional and functional results from using this new experimental platform to yield insights on epigenetic control of the heart. We focus on the multitude of methodologies and workflows needed to quantify responses to HDACis in hiPSC-CMs. This overview can help highlight the power and the limitations of hiPSC-CMs as a scalable experimental model in capturing epigenetic responses relevant to the human heart.
Collapse
Affiliation(s)
| | | | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA; (M.R.P.); (G.W.M.)
| |
Collapse
|
29
|
English JD, Tian S, Wang Z, Luzum JA. Association of Valproic Acid Use With Post-Myocardial Infarction Heart Failure Development: A Meta-Analysis of Two Retrospective Case-Control Studies. J Cardiovasc Pharmacol Ther 2022; 27:10742484221140303. [PMID: 36416392 PMCID: PMC9841513 DOI: 10.1177/10742484221140303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Despite advances in treatments, myocardial infarction (MI) remains a significant cause of morbidity and mortality worldwide. Our team has previously shown that valproic acid (VPA) is cardio-protective when administered to rats post-MI. The aim of this study was to investigate the association of VPA use with post-MI heart failure (HF) development in humans. METHODS This study was a random effects meta-analysis of two retrospective case-control studies collected from electronic health record (Michigan Medicine) and claims data (OptumInsight). Cases with an active prescription for VPA at the time of their MI were matched 1:4 to controls not taking VPA at the time of their MI by multiple demographic and clinical characteristics. The primary outcome, time-to-HF development, was analyzed using the Fine-Gray competing risks model of any VPA prescription versus no VPA prescription. An exploratory analysis was conducted to evaluate the association of different VPA doses (≥1000 mg/day vs <1000 mg/day vs 0 mg/day VPA). RESULTS In total, the datasets included 1313 patients (249 cases and 1064 controls). In the meta-analysis, any dose of VPA during an MI tended to be protective against incident HF post-MI (HR = 0.87; 95% CI = 0.72-1.01). However, when stratified by dose, high-dose VPA (≥1000 mg/day) significantly associated with 30% reduction in risk for HF post-MI (HR = 0.70; 95% CI = 0.49-0.91), whereas low-dose VPA (<1000 mg/day) did not (HR = 0.95; 95% CI = 0.78-1.13). CONCLUSION VPA doses ≥1000 mg/day may provide post-MI cardio-protection resulting in a reduced incidence of HF.
Collapse
Affiliation(s)
- Joseph D English
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Shuo Tian
- Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Wu Y, Xu H, Tu X, Gao Z. The Role of Short-Chain Fatty Acids of Gut Microbiota Origin in Hypertension. Front Microbiol 2021; 12:730809. [PMID: 34650536 PMCID: PMC8506212 DOI: 10.3389/fmicb.2021.730809] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Hypertension is a significant risk factor for cardiovascular and cerebrovascular diseases, and its development involves multiple mechanisms. Gut microbiota has been reported to be closely linked to hypertension. Short-chain fatty acids (SCFAs)-the metabolites of gut microbiota-participate in hypertension development through various pathways, including specific receptors, immune system, autonomic nervous system, metabolic regulation and gene transcription. This article reviews the possible mechanisms of SCFAs in regulating blood pressure and the prospects of SCFAs as a target to prevent and treat hypertension.
Collapse
Affiliation(s)
- Yeshun Wu
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Hongqing Xu
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xiaoming Tu
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Zhenyan Gao
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| |
Collapse
|
31
|
Pedro Ferreira J, Pitt B, Zannad F. Histone deacetylase inhibitors for cardiovascular conditions and healthy longevity. THE LANCET. HEALTHY LONGEVITY 2021; 2:e371-e379. [DOI: 10.1016/s2666-7568(21)00061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/19/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022] Open
|
32
|
Gadasheva Y, Nolze A, Grossmann C. Posttranslational Modifications of the Mineralocorticoid Receptor and Cardiovascular Aging. Front Mol Biosci 2021; 8:667990. [PMID: 34124152 PMCID: PMC8193679 DOI: 10.3389/fmolb.2021.667990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/07/2021] [Indexed: 01/05/2023] Open
Abstract
During aging, the cardiovascular system is especially prone to a decline in function and to life-expectancy limiting diseases. Cardiovascular aging is associated with increased arterial stiffness and vasoconstriction as well as left ventricular hypertrophy and reduced diastolic function. Pathological changes include endothelial dysfunction, atherosclerosis, fibrosis, hypertrophy, inflammation, and changes in micromilieu with increased production of reactive oxygen and nitrogen species. The renin-angiotensin-aldosterone-system is an important mediator of electrolyte and blood pressure homeostasis and a key contributor to pathological remodeling processes of the cardiovascular system. Its effects are partially conveyed by the mineralocorticoid receptor (MR), a ligand-dependent transcription factor, whose activity increases during aging and cardiovascular diseases without correlating changes of its ligand aldosterone. There is growing evidence that the MR can be enzymatically and non-enzymatically modified and that these modifications contribute to ligand-independent modulation of MR activity. Modifications reported so far include phosphorylation, acetylation, ubiquitination, sumoylation and changes induced by nitrosative and oxidative stress. This review focuses on the different posttranslational modifications of the MR, their impact on MR function and degradation and the possible implications for cardiovascular aging and diseases.
Collapse
Affiliation(s)
- Yekatarina Gadasheva
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Alexander Nolze
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
33
|
Francois A, Canella A, Marcho LM, Stratton MS. Protein acetylation in cardiac aging. J Mol Cell Cardiol 2021; 157:90-97. [PMID: 33915138 DOI: 10.1016/j.yjmcc.2021.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/14/2021] [Accepted: 04/21/2021] [Indexed: 11/15/2022]
Abstract
Biological aging is attributed to progressive dysfunction in systems governing genetic and metabolic integrity. At the cellular level, aging is evident by accumulated DNA damage and mutation, reactive oxygen species, alternate lipid and protein modifications, alternate gene expression programs, and mitochondrial dysfunction. These effects sum to drive altered tissue morphology and organ dysfunction. Protein-acylation has emerged as a critical mediator of age-dependent changes in these processes. Despite decades of research focus from academia and industry, heart failure remains a leading cause of death in the United States while the 5 year mortality rate for heart failure remains over 40%. Over 90% of heart failure deaths occur in patients over the age of 65 and heart failure is the leading cause of hospitalization in Medicare beneficiaries. In 1931, Cole and Koch discovered age-dependent accumulation of phosphates in skeletal muscle. These and similar findings provided supporting evidence for, now well accepted, theories linking metabolism and aging. Nearly two decades later, age-associated alterations in biochemical molecules were described in the heart. From these small beginnings, the field has grown substantially in recent years. This growing research focus on cardiac aging has, in part, been driven by advances on multiple public health fronts that allow population level clinical presentation of aging related disorders. It is estimated that by 2030, 25% of the worldwide population will be over the age of 65. This review provides an overview of acetylation-dependent regulation of biological processes related to cardiac aging and introduces emerging non-acetyl, acyl-lysine modifications in cardiac function and aging.
Collapse
Affiliation(s)
- Ashley Francois
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Alessandro Canella
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lynn M Marcho
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Matthew S Stratton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
34
|
Chang C, Yan J, Yao Z, Zhang C, Li X, Mao H. Effects of Mesenchymal Stem Cell-Derived Paracrine Signals and Their Delivery Strategies. Adv Healthc Mater 2021; 10:e2001689. [PMID: 33433956 PMCID: PMC7995150 DOI: 10.1002/adhm.202001689] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/13/2020] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have been widely studied as a versatile cell source for tissue regeneration and remodeling due to their potent bioactivity, which includes modulation of inflammation response, macrophage polarization toward proregenerative lineage, promotion of angiogenesis, and reduction in fibrosis. This review focuses on profiling the effects of paracrine signals of MSCs, commonly referred to as the secretome, and highlighting the various engineering approaches to tune the MSC secretome. Recent advances in biomaterials‐based therapeutic strategies for delivery of MSCs and MSC‐derived secretome in the form of extracellular vesicles are discussed, along with their advantages and challenges.
Collapse
Affiliation(s)
- Calvin Chang
- Department of Biomedical Engineering, School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Jerry Yan
- Department of Biomedical Engineering, School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Zhicheng Yao
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Chi Zhang
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| | - Xiaowei Li
- Mary and Dick Holland Regenerative Medicine Program and Department of Neurological Sciences University of Nebraska Medical Center Omaha NE 68198 USA
| | - Hai‐Quan Mao
- Department of Biomedical Engineering, School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Materials Science and Engineering, Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| |
Collapse
|
35
|
Lamothe J, Khurana S, Tharmalingam S, Williamson C, Byrne CJ, Lees SJ, Khaper N, Kumar A, Tai T. Oxidative Stress Mediates the Fetal Programming of Hypertension by Glucocorticoids. Antioxidants (Basel) 2021; 10:antiox10040531. [PMID: 33805403 PMCID: PMC8066984 DOI: 10.3390/antiox10040531] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
The field of cardiovascular fetal programming has emphasized the importance of the uterine environment on postnatal cardiovascular health. Studies have linked increased fetal glucocorticoid exposure, either from exogenous sources (such as dexamethasone (Dex) injections), or from maternal stress, to the development of adult cardiovascular pathologies. Although the mechanisms are not fully understood, alterations in gene expression driven by altered oxidative stress and epigenetic pathways are implicated in glucocorticoid-mediated cardiovascular programming. Antioxidants, such as the naturally occurring polyphenol epigallocatechin gallate (EGCG), or the superoxide dismutase (SOD) 4-hydroxy-TEMPO (TEMPOL), have shown promise in the prevention of cardiovascular dysfunction and programming. This study investigated maternal antioxidant administration with EGCG or TEMPOL and their ability to attenuate the fetal programming of hypertension via Dex injections in WKY rats. Results from this study indicate that, while Dex-programming increased blood pressure in male and female adult offspring, administration of EGCG or TEMPOL via maternal drinking water attenuated Dex-programmed increases in blood pressure, as well as changes in adrenal mRNA and protein levels of catecholamine biosynthetic enzymes phenylalanine hydroxylase (PAH), tyrosine hydroxylase (TH), dopamine beta hydroxylase (DBH), and phenylethanolamine N-methyltransferase (PNMT), in a sex-specific manner. Furthermore, programmed male offspring displayed reduced antioxidant glutathione peroxidase 1 (Gpx1) expression, increased superoxide dismutase 1 (SOD1) and catalase (CAT) expression, and increased pro-oxidant NADPH oxidase activator 1 (Noxa1) expression in the adrenal glands. In addition, prenatal Dex exposure alters expression of epigenetic regulators histone deacetylase (HDAC) 1, 5, 6, 7, 11, in male and HDAC7 in female offspring. These results suggest that glucocorticoids may mediate the fetal programming of hypertension via alteration of epigenetic machinery and oxidative stress pathways.
Collapse
Affiliation(s)
- Jeremy Lamothe
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (J.L.); (S.T.); (N.K.); (A.K.)
| | - Sandhya Khurana
- Medical Science Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada;
| | - Sujeenthar Tharmalingam
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (J.L.); (S.T.); (N.K.); (A.K.)
- Medical Science Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada;
- Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada; (C.W.); (C.J.B.)
| | - Chad Williamson
- Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada; (C.W.); (C.J.B.)
| | - Collin J. Byrne
- Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada; (C.W.); (C.J.B.)
| | - Simon J. Lees
- Biology, Lakehead University, Thunder Bay, ON P3E 2C6, Canada;
- Medical Science Division, Northern Ontario School of Medicine, Thunder Bay, ON P7B 5E1, Canada
| | - Neelam Khaper
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (J.L.); (S.T.); (N.K.); (A.K.)
- Biology, Lakehead University, Thunder Bay, ON P3E 2C6, Canada;
- Medical Science Division, Northern Ontario School of Medicine, Thunder Bay, ON P7B 5E1, Canada
| | - Aseem Kumar
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (J.L.); (S.T.); (N.K.); (A.K.)
- Medical Science Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada;
- Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada; (C.W.); (C.J.B.)
| | - T.C. Tai
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (J.L.); (S.T.); (N.K.); (A.K.)
- Medical Science Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada;
- Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada; (C.W.); (C.J.B.)
- Correspondence:
| |
Collapse
|
36
|
The Therapeutic Potential of Epigenome-Modifying Drugs in Cardiometabolic Disease. CURRENT GENETIC MEDICINE REPORTS 2021. [DOI: 10.1007/s40142-021-00198-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
37
|
Gutiérrez-Cuevas J, Sandoval-Rodriguez A, Meza-Rios A, Monroy-Ramírez HC, Galicia-Moreno M, García-Bañuelos J, Santos A, Armendariz-Borunda J. Molecular Mechanisms of Obesity-Linked Cardiac Dysfunction: An Up-Date on Current Knowledge. Cells 2021; 10:629. [PMID: 33809061 PMCID: PMC8000147 DOI: 10.3390/cells10030629] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is defined as excessive body fat accumulation, and worldwide obesity has nearly tripled since 1975. Excess of free fatty acids (FFAs) and triglycerides in obese individuals promote ectopic lipid accumulation in the liver, skeletal muscle tissue, and heart, among others, inducing insulin resistance, hypertension, metabolic syndrome, type 2 diabetes (T2D), atherosclerosis, and cardiovascular disease (CVD). These diseases are promoted by visceral white adipocyte tissue (WAT) dysfunction through an increase in pro-inflammatory adipokines, oxidative stress, activation of the renin-angiotensin-aldosterone system (RAAS), and adverse changes in the gut microbiome. In the heart, obesity and T2D induce changes in substrate utilization, tissue metabolism, oxidative stress, and inflammation, leading to myocardial fibrosis and ultimately cardiac dysfunction. Peroxisome proliferator-activated receptors (PPARs) are involved in the regulation of carbohydrate and lipid metabolism, also improve insulin sensitivity, triglyceride levels, inflammation, and oxidative stress. The purpose of this review is to provide an update on the molecular mechanisms involved in obesity-linked CVD pathophysiology, considering pro-inflammatory cytokines, adipokines, and hormones, as well as the role of oxidative stress, inflammation, and PPARs. In addition, cell lines and animal models, biomarkers, gut microbiota dysbiosis, epigenetic modifications, and current therapeutic treatments in CVD associated with obesity are outlined in this paper.
Collapse
Affiliation(s)
- Jorge Gutiérrez-Cuevas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Ana Sandoval-Rodriguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Alejandra Meza-Rios
- Tecnologico de Monterrey, Campus Guadalajara, Zapopan, School of Medicine and Health Sciences, Jalisco 45201, Mexico; (A.M.-R.); (A.S.)
| | - Hugo Christian Monroy-Ramírez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Marina Galicia-Moreno
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Jesús García-Bañuelos
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Arturo Santos
- Tecnologico de Monterrey, Campus Guadalajara, Zapopan, School of Medicine and Health Sciences, Jalisco 45201, Mexico; (A.M.-R.); (A.S.)
| | - Juan Armendariz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
- Tecnologico de Monterrey, Campus Guadalajara, Zapopan, School of Medicine and Health Sciences, Jalisco 45201, Mexico; (A.M.-R.); (A.S.)
| |
Collapse
|
38
|
Soranno DE, Kirkbride-Romeo L, Wennersten SA, Ding K, Cavasin MA, Baker P, Altmann C, Bagchi RA, Haefner KR, Steinkühler C, Montford JR, Keith B, Gist KM, McKinsey TA, Faubel S. Acute Kidney Injury Results in Long-Term Diastolic Dysfunction That Is Prevented by Histone Deacetylase Inhibition. ACTA ACUST UNITED AC 2021; 6:119-133. [PMID: 33665513 PMCID: PMC7907538 DOI: 10.1016/j.jacbts.2020.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 01/06/2023]
Abstract
This is the first long-term (1-year) study to evaluate both the kidney and systemic sequelae of acute kidney injury in mice. Serial kidney function was measured via transcutaneous glomerular filtration rate. AKI resulted in diastolic dysfunction, followed by hypertension. Ejection fraction was preserved. One year after AKI, cardiac ATP levels were reduced compared with sham controls. Mice treated with the histone deacetylase inhibitor, ITF2357, maintained normal diastolic function normal blood pressure, and normal cardiac ATP after AKI. Metabolomics data suggest that treatment with ITF2357 preserves pathways related to energy metabolism.
Growing epidemiological data demonstrate that acute kidney injury (AKI) is associated with long-term cardiovascular morbidity and mortality. Here, the authors present a 1-year study of cardiorenal outcomes following bilateral ischemia-reperfusion injury in male mice. These data suggest that AKI causes long-term dysfunction in the cardiac metabolome, which is associated with diastolic dysfunction and hypertension. Mice treated with the histone deacetylase inhibitor, ITF2357, had preservation of cardiac function and remained normotensive throughout the study. ITF2357 did not protect against the development of kidney fibrosis after AKI.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, Aurora, Colorado, USA.,Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lara Kirkbride-Romeo
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, Aurora, Colorado, USA
| | - Sara A Wennersten
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Kathy Ding
- Department of Pediatrics, Pediatric Nephrology, University of Colorado, Aurora, Colorado, USA
| | - Maria A Cavasin
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Peter Baker
- Department of Pediatrics, Clinical Genetics and Metabolism, University of Colorado, Aurora, Colorado, USA
| | - Christopher Altmann
- Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA
| | - Rushita A Bagchi
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Korey R Haefner
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | | | - John R Montford
- Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| | - Brysen Keith
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katja M Gist
- Department of Pediatrics, Pediatric Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Timothy A McKinsey
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado, Aurora, Colorado, USA
| | - Sarah Faubel
- Department of Medicine, Division of Renal Disease and Hypertension, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
39
|
Chun P. Therapeutic effects of histone deacetylase inhibitors on heart disease. Arch Pharm Res 2020; 43:1276-1296. [PMID: 33245518 DOI: 10.1007/s12272-020-01297-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/22/2020] [Indexed: 01/04/2023]
Abstract
A wide range of histone deacetylase (HDAC) inhibitors have been studied for their therapeutic potential because the excessive activity and expression of HDACs have been implicated in the pathogenesis of cardiac diseases. An increasing number of preclinical studies have demonstrated the cardioprotective effects of numerous HDAC inhibitors, suggesting a wide variety of mechanisms by which the inhibitors protect against cardiac stress, such as the suppression of cardiac fibrosis and fetal gene expression, enhancement of angiogenesis and mitochondrial biogenesis, prevention of electrical remodeling, and regulation of apoptosis, autophagy, and cell cycle arrest. For the development of isoform-selective HDAC inhibitors with high efficacy and low toxicity, it is important to identify and understand the mechanisms responsible for the effects of the inhibitors. This review highlights the preclinical effects of HDAC inhibitors that act against Zn2+-dependent HDACs and the underlying mechanisms of their protective effects against cardiac hypertrophy, hypertension, myocardial infarction, heart failure, and atrial fibrillation.
Collapse
Affiliation(s)
- Pusoon Chun
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Inje-ro, Gimhae, Gyeongnam, 50834, Republic of Korea.
| |
Collapse
|
40
|
Protective Effect of Vitis labrusca Leaves Extract on Cardiovascular Dysfunction through HMGB1-TLR4-NFκB Signaling in Spontaneously Hypertensive Rats. Nutrients 2020; 12:nu12103096. [PMID: 33050676 PMCID: PMC7601160 DOI: 10.3390/nu12103096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
The Vitis labrusca is a grapevine that has antioxidant, neuroprotective, hepatoprotective, and anticarcinogenic activity. However, the effect of Vitis labrusca leaves on the cardiovascular system is yet to be ascertained. The present study was designed to investigate the effects of Vitis labrusca leaves extract (HP1) on cardiovascular remodeling in spontaneously hypertensive rats. Experiments were performed in rats and were randomly divided into the following groups: Wistar Kyoto rat (WKY), normal control group; spontaneously hypertensive rats (SHR), negative control group; SHR + Losa, positive control group (losartan, 10 mg/kg/daily, AT1 receptor blocker) and SHR + HP1 (100 mg/kg/daily). HP1 was orally administered daily for 4 weeks. The HP1 treatment significantly improved blood pressure, electrocardiographic parameters, and echocardiogram parameters compared to hypertensive rats. Additionally, the left ventricular (LV) remodeling and LV dysfunction were significantly improved in HP1-treated hypertensive rats. Furthermore, an increase in fibrotic area has been observed in hypertensive rats compared with WKY. However, administration of HP1 significantly attenuated cardiac fibrosis in hypertensive rats. Moreover, HP1 suppressed the expression of high mobility group box 1 (HMGB1), toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), receptor for advanced glycation end products (RAGE), and extracellular signal-regulated kinases (ERK1/2) induced by hypertensive rats, resulting in improved vascular remodeling. Therefore, these results suggest that HP1 can improve the cardiovascular remodeling in hypertensive rats, and the mechanisms may be related to the suppressive effect of HP1 on HMGB1-TLR4-NFκB signaling in the cardiovascular system. Thus, the protective role of the traditional herbal medicine HP1 may provide new insights into the development of therapeutic drugs on the development of hypertensive cardiovascular dysfunction.
Collapse
|
41
|
Verhaar BJH, Prodan A, Nieuwdorp M, Muller M. Gut Microbiota in Hypertension and Atherosclerosis: A Review. Nutrients 2020; 12:E2982. [PMID: 33003455 PMCID: PMC7601560 DOI: 10.3390/nu12102982] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/22/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota and its metabolites such as short chain fatty acids (SCFA), lipopolysaccharides (LPS), and trimethylamine-N-oxide (TMAO) impact cardiovascular health. In this review, we discuss how gut microbiota and gut metabolites can affect hypertension and atherosclerosis. Hypertensive patients were shown to have lower alpha diversity, lower abundance of SCFA-producing microbiota, and higher abundance of gram-negative bacteria, which are a source of LPS. Animal studies point towards a direct role for SCFAs in blood pressure regulation and show that LPS has pro-inflammatory effects. Translocation of LPS into the systemic circulation is a consequence of increased gut permeability. Atherosclerosis, a multifactorial disease, is influenced by the gut microbiota through multiple pathways. Many studies have focused on the pro-atherogenic role of TMAO, however, it is not clear if this is a causal factor. In addition, gut microbiota play a key role in bile acid metabolism and some interventions targeting bile acid receptors tend to decrease atherosclerosis. Concluding, gut microbiota affect hypertension and atherosclerosis through many pathways, providing a wide range of potential therapeutic targets. Challenges ahead include translation of findings and mechanisms to humans and development of therapeutic interventions that target cardiovascular risk by modulation of gut microbes and metabolites.
Collapse
Affiliation(s)
- Barbara J. H. Verhaar
- Department of Internal Medicine, Section Geriatrics, Amsterdam Cardiovascular Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, 1011-1109 Amsterdam, The Netherlands;
- Department of Internal Medicine, Section Vascular Medicine, Universiteit van Amsterdam, Amsterdam UMC, 1011-1109 Amsterdam, The Netherlands; (A.P.); (M.N.)
| | - Andrei Prodan
- Department of Internal Medicine, Section Vascular Medicine, Universiteit van Amsterdam, Amsterdam UMC, 1011-1109 Amsterdam, The Netherlands; (A.P.); (M.N.)
| | - Max Nieuwdorp
- Department of Internal Medicine, Section Vascular Medicine, Universiteit van Amsterdam, Amsterdam UMC, 1011-1109 Amsterdam, The Netherlands; (A.P.); (M.N.)
| | - Majon Muller
- Department of Internal Medicine, Section Geriatrics, Amsterdam Cardiovascular Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, 1011-1109 Amsterdam, The Netherlands;
| |
Collapse
|
42
|
The Role of DNMT and HDACs in the Fetal Programming of Hypertension by Glucocorticoids. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5751768. [PMID: 32318239 PMCID: PMC7149440 DOI: 10.1155/2020/5751768] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/07/2020] [Indexed: 12/13/2022]
Abstract
The causes of hypertension are complex and involve both genetic and environmental factors. Environment changes during fetal development have been linked to adult diseases including hypertension. Studies show that timed in utero exposure to the synthetic glucocorticoid (GC) dexamethasone (Dex) results in the development of hypertension in adult rats. Evidence suggests that in utero stress can alter patterns of gene expression, possibly a result of alterations in the topology of the genome by epigenetic markers such as DNA methyltransferases (DNMTs) and histone deacetylases (HDACs). The objective of this study was to determine the effects of epigenetic regulators in the fetal programming and the development of adult hypertension. Specifically, this research examined the effects of the HDAC inhibitor valproic acid (VPA) and the DNMT inhibitor 5-aza-2′-deoxycytidine (5aza2DC) on blood pressure (BP) and gene expression in prenatal Dex-programmed rats. Data suggest that both VPA and 5aza2DC attenuated the Dex-mediated development of hypertension and restored BP to control levels. Epigenetic DNMT inhibition (DNMTi) or HDAC inhibition (HDACi) also successfully attenuated elevations in the majority of altered catecholamine (CA) enzyme expression, phenylethanolamine N-methyltransferase (PNMT) protein, and elevated epinephrine (Epi) levels in males. Although females responded to HDACi similar to males, DNMTi drove increased glucocorticoid receptor (GR) and PNMT expression and elevations in circulating Epi in females despite showing normotensive BP.
Collapse
|
43
|
Effects of third-generation β-blockers, atenolol or amlodipine on blood pressure variability and target organ damage in spontaneously hypertensive rats. J Hypertens 2020; 38:536-545. [DOI: 10.1097/hjh.0000000000002284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Role of HDACs in cardiac electropathology: Therapeutic implications for atrial fibrillation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118459. [DOI: 10.1016/j.bbamcr.2019.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022]
|
45
|
Palomo M, Vera M, Martin S, Torramadé‐Moix S, Martinez‐Sanchez J, Belen Moreno A, Carreras E, Escolar G, Cases A, Díaz‐Ricart M. Up-regulation of HDACs, a harbinger of uraemic endothelial dysfunction, is prevented by defibrotide. J Cell Mol Med 2020; 24:1713-1723. [PMID: 31782253 PMCID: PMC6991634 DOI: 10.1111/jcmm.14865] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/24/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Endothelial dysfunction is an earlier contributor to the development of atherosclerosis in chronic kidney disease (CKD), in which the role of epigenetic triggers cannot be ruled out. Endothelial protective strategies, such as defibrotide (DF), may be useful in this scenario. We evaluated changes induced by CKD on endothelial cell proteome and explored the effect of DF and the mechanisms involved. Human umbilical cord vein endothelial cells were exposed to sera from healthy donors (n = 20) and patients with end-stage renal disease on haemodialysis (n = 20). Differential protein expression was investigated by using a proteomic approach, Western blot and immunofluorescence. HDAC1 and HDAC2 overexpression was detected. Increased HDAC1 expression occurred at both cytoplasm and nucleus. These effects were dose-dependently inhibited by DF. Both the HDACs inhibitor trichostatin A and DF prevented the up-regulation of the endothelial dysfunction markers induced by the uraemic milieu: intercellular adhesion molecule-1, surface Toll-like receptor-4, von Willebrand Factor and reactive oxygen species. Moreover, DF down-regulated HDACs expression through the PI3/AKT signalling pathway. HDACs appear as key modulators of the CKD-induced endothelial dysfunction as specific blockade by trichostatin A or by DF prevents endothelial dysfunction responses to the CKD insult. Moreover, DF exerts its endothelial protective effect by inhibiting HDAC up-regulation likely through PI3K/AKT.
Collapse
Affiliation(s)
- Marta Palomo
- HematopathologyCentre Diagnòstic Biomèdic (CDB)Hospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
- Josep Carreras Leukaemia Research InstituteHospital Clinic/University of Barcelona CampusBarcelonaSpain
- Barcelona Endothelium Team (BET)BarcelonaSpain
| | - Manel Vera
- Nephrology DepartmentHospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
| | - Susana Martin
- HematopathologyCentre Diagnòstic Biomèdic (CDB)Hospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
| | - Sergi Torramadé‐Moix
- HematopathologyCentre Diagnòstic Biomèdic (CDB)Hospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
| | - Julia Martinez‐Sanchez
- HematopathologyCentre Diagnòstic Biomèdic (CDB)Hospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
- Josep Carreras Leukaemia Research InstituteHospital Clinic/University of Barcelona CampusBarcelonaSpain
- Barcelona Endothelium Team (BET)BarcelonaSpain
| | - Ana Belen Moreno
- HematopathologyCentre Diagnòstic Biomèdic (CDB)Hospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
| | - Enric Carreras
- Josep Carreras Leukaemia Research InstituteHospital Clinic/University of Barcelona CampusBarcelonaSpain
- Barcelona Endothelium Team (BET)BarcelonaSpain
| | - Ginés Escolar
- HematopathologyCentre Diagnòstic Biomèdic (CDB)Hospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
| | - Aleix Cases
- Nephrology DepartmentHospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
| | - Maribel Díaz‐Ricart
- HematopathologyCentre Diagnòstic Biomèdic (CDB)Hospital ClinicInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de Barcelona (UB)BarcelonaSpain
- Barcelona Endothelium Team (BET)BarcelonaSpain
| |
Collapse
|
46
|
El‐Naggar AM, Somasekharan SP, Wang Y, Cheng H, Negri GL, Pan M, Wang XQ, Delaidelli A, Rafn B, Cran J, Zhang F, Zhang H, Colborne S, Gleave M, Mandinova A, Kedersha N, Hughes CS, Surdez D, Delattre O, Wang Y, Huntsman DG, Morin GB, Sorensen PH. Class I HDAC inhibitors enhance YB-1 acetylation and oxidative stress to block sarcoma metastasis. EMBO Rep 2019; 20:e48375. [PMID: 31668005 PMCID: PMC6893361 DOI: 10.15252/embr.201948375] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/29/2019] [Accepted: 10/06/2019] [Indexed: 12/13/2022] Open
Abstract
Outcomes for metastatic Ewing sarcoma and osteosarcoma are dismal and have not changed for decades. Oxidative stress attenuates melanoma metastasis, and melanoma cells must reduce oxidative stress to metastasize. We explored this in sarcomas by screening for oxidative stress sensitizers, which identified the class I HDAC inhibitor MS-275 as enhancing vulnerability to reactive oxygen species (ROS) in sarcoma cells. Mechanistically, MS-275 inhibits YB-1 deacetylation, decreasing its binding to 5'-UTRs of NFE2L2 encoding the antioxidant factor NRF2, thereby reducing NFE2L2 translation and synthesis of NRF2 to increase cellular ROS. By global acetylomics, MS-275 promotes rapid acetylation of the YB-1 RNA-binding protein at lysine-81, blocking binding and translational activation of NFE2L2, as well as known YB-1 mRNA targets, HIF1A, and the stress granule nucleator, G3BP1. MS-275 dramatically reduces sarcoma metastasis in vivo, but an MS-275-resistant YB-1K81-to-alanine mutant restores metastatic capacity and NRF2, HIF1α, and G3BP1 synthesis in MS-275-treated mice. These studies describe a novel function for MS-275 through enhanced YB-1 acetylation, thus inhibiting YB-1 translational control of key cytoprotective factors and its pro-metastatic activity.
Collapse
Affiliation(s)
- Amal M El‐Naggar
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
- Department of PathologyFaculty of MedicineMenoufia UniversityShibin El KomEgypt
| | | | - Yemin Wang
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | | | | | - Melvin Pan
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Xue Qi Wang
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Alberto Delaidelli
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Bo Rafn
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Jordan Cran
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Fan Zhang
- Vancouver Prostate CentreVancouverBCCanada
| | - Haifeng Zhang
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | | | | | - Anna Mandinova
- Brigham and Women's HospitalHarvard UniversityBostonMAUSA
| | - Nancy Kedersha
- Massachusetts General HospitalHarvard UniversityBostonMAUSA
| | - Christopher S Hughes
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | | | | | | | - David G Huntsman
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| | - Gregg B Morin
- Michael Smith Genome Sciences CentreVancouverBCCanada
| | - Poul H Sorensen
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Molecular Oncology, BC Cancerpart of the Provincial Health Services AuthorityVancouverBCCanada
| |
Collapse
|
47
|
CD47 Deficiency Attenuates Isoproterenol-Induced Cardiac Remodeling in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7121763. [PMID: 31827695 PMCID: PMC6885801 DOI: 10.1155/2019/7121763] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022]
Abstract
In this study, we investigated whether CD47 deficiency attenuates isoproterenol- (ISO-) induced cardiac remodeling in mice. Cardiac remodeling was induced by intraperitoneal (i.p.) injection of ISO (60 mg·kg−1·d−1 in 100 μl of sterile normal saline) daily for 14 days and was confirmed by increased levels of lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB), increased heart weight to body weight (HW/BW) ratios, and visible cardiac fibrosis. Apoptosis was evaluated by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. Levels of malondialdehyde (MDA) and reactive oxygen species (ROS) were found to be significantly higher in the ISO group than in the control group, while superoxide dismutase (SOD) levels were suppressed in the ISO group. However, CD47 knockout significantly limited ISO-induced increases in LDH, CK-MB, and HW/BW ratios, cardiac fibrosis, oxidative stress, and apoptosis in the heart. In addition, CD47 deficiency also increased p-AMPK and LAMP2 expression and decreased HDAC3, cleaved Caspase-3, cleaved Caspase-9, LC3II, and p62 expression in cardiac tissues. In conclusion, CD47 deficiency reduced i.p. ISO-induced cardiac remodeling probably by inhibiting the HDAC3 pathway, improving AMPK signaling and autophagy flux, and rescuing autophagic clearance.
Collapse
|
48
|
Zolotareva O, Saik OV, Königs C, Bragina EY, Goncharova IA, Freidin MB, Dosenko VE, Ivanisenko VA, Hofestädt R. Comorbidity of asthma and hypertension may be mediated by shared genetic dysregulation and drug side effects. Sci Rep 2019; 9:16302. [PMID: 31705029 PMCID: PMC6841742 DOI: 10.1038/s41598-019-52762-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Asthma and hypertension are complex diseases coinciding more frequently than expected by chance. Unraveling the mechanisms of comorbidity of asthma and hypertension is necessary for choosing the most appropriate treatment plan for patients with this comorbidity. Since both diseases have a strong genetic component in this article we aimed to find and study genes simultaneously associated with asthma and hypertension. We identified 330 shared genes and found that they form six modules on the interaction network. A strong overlap between genes associated with asthma and hypertension was found on the level of eQTL regulated genes and between targets of drugs relevant for asthma and hypertension. This suggests that the phenomenon of comorbidity of asthma and hypertension may be explained by altered genetic regulation or result from drug side effects. In this work we also demonstrate that not only drug indications but also contraindications provide an important source of molecular evidence helpful to uncover disease mechanisms. These findings give a clue to the possible mechanisms of comorbidity and highlight the direction for future research.
Collapse
Affiliation(s)
- Olga Zolotareva
- Bielefeld University, International Research Training Group "Computational Methods for the Analysis of the Diversity and Dynamics of Genomes" and Genome Informatics, Faculty of Technology and Center for Biotechnology, Bielefeld, Germany.
| | - Olga V Saik
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Cassandra Königs
- Bielefeld University, Bioinformatics and Medical Informatics Department, Bielefeld, Germany
| | - Elena Yu Bragina
- Research Institute of Medical Genetics, Tomsk NRMC, Tomsk, Russia
| | | | - Maxim B Freidin
- Research Institute of Medical Genetics, Tomsk NRMC, Tomsk, Russia
| | | | - Vladimir A Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Ralf Hofestädt
- Bielefeld University, Bioinformatics and Medical Informatics Department, Bielefeld, Germany
| |
Collapse
|
49
|
Abstract
Supplemental Digital Content is available in the text. If unifying principles could be revealed for how the same genome encodes different eukaryotic cells and for how genetic variability and environmental input are integrated to impact cardiovascular health, grand challenges in basic cell biology and translational medicine may succumb to experimental dissection. A rich body of work in model systems has implicated chromatin-modifying enzymes, DNA methylation, noncoding RNAs, and other transcriptome-shaping factors in adult health and in the development, progression, and mitigation of cardiovascular disease. Meanwhile, deployment of epigenomic tools, powered by next-generation sequencing technologies in cardiovascular models and human populations, has enabled description of epigenomic landscapes underpinning cellular function in the cardiovascular system. This essay aims to unpack the conceptual framework in which epigenomes are studied and to stimulate discussion on how principles of chromatin function may inform investigations of cardiovascular disease and the development of new therapies.
Collapse
Affiliation(s)
- Manuel Rosa-Garrido
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Douglas J Chapski
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Thomas M Vondriska
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles.
| |
Collapse
|
50
|
CaMKII Activity in the Inflammatory Response of Cardiac Diseases. Int J Mol Sci 2019; 20:ijms20184374. [PMID: 31489895 PMCID: PMC6770001 DOI: 10.3390/ijms20184374] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022] Open
Abstract
Inflammation is a physiological process by which the body responds to external insults and stress conditions, and it is characterized by the production of pro-inflammatory mediators such as cytokines. The acute inflammatory response is solved by removing the threat. Conversely, a chronic inflammatory state is established due to a prolonged inflammatory response and may lead to tissue damage. Based on the evidence of a reciprocal regulation between inflammation process and calcium unbalance, here we described the involvement of a calcium sensor in cardiac diseases with inflammatory drift. Indeed, the Ca2+/calmodulin-dependent protein kinase II (CaMKII) is activated in several diseases with an inflammatory component, such as myocardial infarction, ischemia/reperfusion injury, pressure overload/hypertrophy, and arrhythmic syndromes, in which it actively regulates pro-inflammatory signaling, among which includes nuclear factor kappa-B (NF-κB), thus contributing to pathological cardiac remodeling. Thus, CaMKII may represent a key target to modulate the severity of the inflammatory-driven degeneration.
Collapse
|