1
|
Yang L, Li X, Ni L, Lin Y. Treatment of endothelial cell dysfunction in atherosclerosis: a new perspective integrating traditional and modern approaches. Front Physiol 2025; 16:1555118. [PMID: 40206381 PMCID: PMC11979162 DOI: 10.3389/fphys.2025.1555118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Atherosclerosis (AS), a prime causative factor in cardiovascular disease, originates from endothelial cell dysfunction (ECD). Comprising a vital part of the vascular endothelium, endothelial cells play a crucial role in maintaining vascular homeostasis, optimizing redox balance, and regulating inflammatory responses. More evidence shows that ECD not only serves as an early harbinger of AS but also exhibits a strong association with disease progression. In recent years, the treatment strategies for ECD have been continuously evolving, encompassing interventions ranging from lifestyle modifications to traditional pharmacotherapy aimed at reducing risk factors, which also have demonstrated the ability to improve endothelial cell function. Additionally, novel strategies such as promising biotherapy and gene therapy have drawn attention. These methods have demonstrated enormous potential and promising prospects in improving endothelial function and reversing AS. However, it is essential to remain cognizant that the current treatments still present significant challenges regarding therapeutic efficacy, long-term safety, and ethical issues. This article aims to provide a systematic review of these treatment methods, analyze the mechanisms and efficacy of various therapeutic strategies, with the goal of offering insights and guidance for clinical practice, and further advancing the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
2
|
Ortega-Hernández S, González-Sosa S, Conde-Martel A, Trullàs JC, Llàcer P, Pérez-Silvestre J, Arévalo-Lorido JC, Casado J, Formiga F, Manzano L, Lorenzo-Villalba N, Montero-Pérez-Barquero M. Prognostic Impact of Statins in Heart Failure with Preserved Ejection Fraction. J Clin Med 2024; 13:5844. [PMID: 39407904 PMCID: PMC11477314 DOI: 10.3390/jcm13195844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Heart failure (HF) with preserved ejection fraction (pEF) has lacked effective treatments for reducing mortality. However, previous studies have found an association between statin use and decreased mortality in patients with HFpEF. The aim of this study was to analyse whether statin therapy is associated with a reduction in mortality in these patients and whether the effect differs according to the presence or absence of ischaemic heart disease (IHD). Methods: We analysed data from the National Registry of Heart Failure, a prospective study that included patients admitted for HF in Internal Medicine units nationwide. Patients with HFpEF were classified according to the use of statins, and the differences between the two groups were analysed. A multivariable analysis was performed using Cox regression to assess factors independently related to mortality. Results: A total of 2788 patients with HFpEF were included; 63% of them were women with a mean age of 80.1 (±7.8) years. The statin-treated group (40.2%) was younger, with better functional status, and had a more common diagnosis of vascular disease and lower frequency of atrial fibrillation. The most frequent aetiology of HF in both groups was the hypertensive one. Nevertheless, ischaemic HF was more common in those who received statins (24.8% vs. 9.6%; p < 0.001). Multivariable analysis showed lower mortality at the 1-year follow-up in statin-treated patients (OR: 0.74; 95%CI: 0.61-0.89; p = 0.002). This association was observed in patients without IHD (p < 0.001) but not in those with IHD (p = 0.11). Conclusions: Statins are associated with a decrease in total mortality in patients with HFpEF. This benefit occurs mainly in those without IHD.
Collapse
Affiliation(s)
- Samanta Ortega-Hernández
- Internal Medicine Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Las Palmas, Spain; (S.O.-H.)
| | - Sonia González-Sosa
- Internal Medicine Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Las Palmas, Spain; (S.O.-H.)
- Health Sciences Faculty, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas, Spain
| | - Alicia Conde-Martel
- Internal Medicine Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Las Palmas, Spain; (S.O.-H.)
- Health Sciences Faculty, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas, Spain
| | - Joan Carles Trullàs
- Internal Medicine Department, Hospital d’Olot i Comarcal de la Garrotxa, 17800 Girona, Spain;
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IrisCC), Faculty of Medicine, University of Vic—Central University of Catalonia (UVic-UCC), Ctra. de Roda, 70, 08500 Vic, Barcelona, Spain
| | - Pau Llàcer
- Internal Medicine Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Universidad de Alcalá, 28801 Madrid, Spain
| | - José Pérez-Silvestre
- Internal Medicine Department, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain
| | | | - Jesús Casado
- Internal Medicine Department, Hospital Universitario de Getafe, 28905 Madrid, Spain
- Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Francesc Formiga
- Internal Medicine Department, Hospital Universitario Bellvitge, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Luis Manzano
- Internal Medicine Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Universidad de Alcalá, 28801 Madrid, Spain
| | - Noel Lorenzo-Villalba
- Service de Médecine Interne, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| | | |
Collapse
|
3
|
Feng R, Zhang Z, Fan Q. Carbohydrate antigen 125 in congestive heart failure: ready for clinical application? Front Oncol 2023; 13:1161723. [PMID: 38023127 PMCID: PMC10644389 DOI: 10.3389/fonc.2023.1161723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Congestion is the permanent mechanism driving disease progression in patients with acute heart failure (AHF) and also is an important treatment target. However, distinguishing between the two different phenotypes (intravascular congestion and tissue congestion) for personalized treatment remains challenging. Historically, carbohydrate antigen 125 (CA125) has been a frequently used biomarker for the screening, diagnosis, and prognosis of ovarian cancer. Interestingly, CA125 is highly sensitive to tissue congestion and shows potential for clinical monitoring and optimal treatment of congestive heart failure (HF). Furthermore, in terms of right heart function parameters, CA125 levels are more advantageous than other biomarkers of HF. CA125 is expected to become a new biological alternative marker for congestive HF and thereby is expected be widely used in clinical practice.
Collapse
Affiliation(s)
- Rui Feng
- Department of Laboratory Medicine, Wuhan Asian Heart Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Zhenlu Zhang
- Department of Laboratory Medicine, Wuhan Asian Heart Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Qingkun Fan
- Department of Laboratory Medicine, Wuhan Asian Heart Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Ravassa S, López B, Treibel TA, San José G, Losada-Fuentenebro B, Tapia L, Bayés-Genís A, Díez J, González A. Cardiac Fibrosis in heart failure: Focus on non-invasive diagnosis and emerging therapeutic strategies. Mol Aspects Med 2023; 93:101194. [PMID: 37384998 DOI: 10.1016/j.mam.2023.101194] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
Heart failure is a leading cause of mortality and hospitalization worldwide. Cardiac fibrosis, resulting from the excessive deposition of collagen fibers, is a common feature across the spectrum of conditions converging in heart failure. Eventually, either reparative or reactive in nature, in the long-term cardiac fibrosis contributes to heart failure development and progression and is associated with poor clinical outcomes. Despite this, specific cardiac antifibrotic therapies are lacking, making cardiac fibrosis an urgent unmet medical need. In this context, a better patient phenotyping is needed to characterize the heterogenous features of cardiac fibrosis to advance toward its personalized management. In this review, we will describe the different phenotypes associated with cardiac fibrosis in heart failure and we will focus on the potential usefulness of imaging techniques and circulating biomarkers for the non-invasive characterization and phenotyping of this condition and for tracking its clinical impact. We will also recapitulate the cardiac antifibrotic effects of existing heart failure and non-heart failure drugs and we will discuss potential strategies under preclinical development targeting the activation of cardiac fibroblasts at different levels, as well as targeting additional extracardiac processes.
Collapse
Affiliation(s)
- Susana Ravassa
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Begoña López
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Thomas A Treibel
- Institute of Cardiovascular Science, University College London, UK; Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Gorka San José
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Blanca Losada-Fuentenebro
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Leire Tapia
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Antoni Bayés-Genís
- CIBERCV, Carlos III Institute of Health, Madrid, Spain; Servei de Cardiologia i Unitat d'Insuficiència Cardíaca, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Badalona, Spain
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| |
Collapse
|
5
|
González-Herrera F, Clayton NS, Guzmán-Rivera D, Carrillo I, Castillo C, Catalán M, Anfossi R, Quintero-Pertuz H, Quilaqueo ME, Olea-Azar C, Rivera-Meza M, Kemmerling U, Ridley AJ, Vivar R, Maya JD. Statins change the cytokine profile in Trypanosoma cruzi-infected U937 macrophages and murine cardiac tissue through Rho-associated kinases inhibition. Front Immunol 2023; 13:1035589. [PMID: 36713380 PMCID: PMC9874148 DOI: 10.3389/fimmu.2022.1035589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Chronic Chagasic cardiomyopathy (CCC), caused by the protozoan Trypanosoma cruzi, is the most severe manifestation of Chagas disease.CCC is characterized by cardiac inflammation and fibrosis caused by a persistent inflammatory response. Following infection, macrophages secrete inflammatory mediators such as IL-1β, IL-6, and TNF-α to control parasitemia. Although this response contains parasite infection, it causes damage to the heart tissue. Thus, the use of immunomodulators is a rational alternative to CCC. Rho-associated kinase (ROCK) 1 and 2 are RhoA-activated serine/threonine kinases that regulate the actomyosin cytoskeleton. Both ROCKs have been implicated in the polarization of macrophages towards an M1 (pro-inflammatory) phenotype. Statins are FDA-approved lipid-lowering drugs that reduce RhoA signaling by inhibiting geranylgeranyl pyrophosphate (GGPP) synthesis. This work aims to identify the effect of statins on U937 macrophage polarization and cardiac tissue inflammation and its relationship with ROCK activity during T. cruzi infection. Methods PMA-induced, wild-type, GFP-, CA-ROCK1- and CA-ROCK2-expressing U937 macrophages were incubated with atorvastatin, or the inhibitors Y-27632, JSH-23, TAK-242, or C3 exoenzyme incubated with or without T. cruzi trypomastigotes for 30 min to evaluate the activity of ROCK and the M1 and M2 cytokine expression and secretion profiling. Also, ROCK activity was determined in T. cruzi-infected, BALB/c mice hearts. Results In this study, we demonstrate for the first time in macrophages that incubation with T. cruzi leads to ROCK activation via the TLR4 pathway, which triggers NF-κB activation. Inhibition of ROCKs by Y-27632 prevents NF-κB activation and the expression and secretion of M1 markers, as does treatment with atorvastatin. Furthermore, we show that the effect of atorvastatin on the NF-kB pathway and cytokine secretion is mediated by ROCK. Finally, statin treatment decreased ROCK activation and expression, and the pro-inflammatory cytokine production, promoting anti-inflammatory cytokine expression in chronic chagasic mice hearts. Conclusion These results suggest that the statin modulation of the inflammatory response due to ROCK inhibition is a potential pharmacological strategy to prevent cardiac inflammation in CCC.
Collapse
Affiliation(s)
- Fabiola González-Herrera
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Natasha S. Clayton
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Daniela Guzmán-Rivera
- Escuela de Farmacia, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Ileana Carrillo
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Christian Castillo
- Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago, Chile
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Renatto Anfossi
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Helena Quintero-Pertuz
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María Elena Quilaqueo
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Claudio Olea-Azar
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Mario Rivera-Meza
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Integrative Biology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Anne J. Ridley
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Raúl Vivar
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile,*Correspondence: Juan Diego Maya, ; Raúl Vivar,
| | - Juan Diego Maya
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile,*Correspondence: Juan Diego Maya, ; Raúl Vivar,
| |
Collapse
|
6
|
Li Z, Zhao H, Wang J. Metabolism and Chronic Inflammation: The Links Between Chronic Heart Failure and Comorbidities. Front Cardiovasc Med 2021; 8:650278. [PMID: 34026868 PMCID: PMC8131678 DOI: 10.3389/fcvm.2021.650278] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) patients often suffer from multiple comorbidities, such as diabetes, atrial fibrillation, depression, chronic obstructive pulmonary disease, and chronic kidney disease. The coexistance of comorbidities usually leads to multi morbidity and poor prognosis. Treatments for HF patients with multi morbidity are still an unmet clinical need, and finding an effective therapy strategy is of great value. HF can lead to comorbidity, and in return, comorbidity may promote the progression of HF, creating a vicious cycle. This reciprocal correlation indicates there may be some common causes and biological mechanisms. Metabolism remodeling and chronic inflammation play a vital role in the pathophysiological processes of HF and comorbidities, indicating metabolism and inflammation may be the links between HF and comorbidities. In this review, we comprehensively discuss the major underlying mechanisms and therapeutic implications for comorbidities of HF. We first summarize the potential role of metabolism and inflammation in HF. Then, we give an overview of the linkage between common comorbidities and HF, from the perspective of epidemiological evidence to the underlying metabolism and inflammation mechanisms. Moreover, with the help of bioinformatics, we summarize the shared risk factors, signal pathways, and therapeutic targets between HF and comorbidities. Metabolic syndrome, aging, deleterious lifestyles (sedentary behavior, poor dietary patterns, smoking, etc.), and other risk factors common to HF and comorbidities are all associated with common mechanisms. Impaired mitochondrial biogenesis, autophagy, insulin resistance, and oxidative stress, are among the major mechanisms of both HF and comorbidities. Gene enrichment analysis showed the PI3K/AKT pathway may probably play a central role in multi morbidity. Additionally, drug targets common to HF and several common comorbidities were found by network analysis. Such analysis has already been instrumental in drug repurposing to treat HF and comorbidity. And the result suggests sodium-glucose transporter-2 (SGLT-2) inhibitors, IL-1β inhibitors, and metformin may be promising drugs for repurposing to treat multi morbidity. We propose that targeting the metabolic and inflammatory pathways that are common to HF and comorbidities may provide a promising therapeutic strategy.
Collapse
Affiliation(s)
- Zhiwei Li
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Hongmei Zhao
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Abstract
Diffuse myocardial fibrosis resulting from the excessive deposition of collagen fibres through the entire myocardium is encountered in a number of chronic cardiac diseases. This lesion results from alterations in the regulation of fibrillary collagen turnover by fibroblasts, facilitating the excessive deposition of type I and type III collagen fibres within the myocardial interstitium and around intramyocardial vessels. The available evidence suggests that, beyond the extent of fibrous deposits, collagen composition and the physicochemical properties of the fibres are also relevant in the detrimental effects of diffuse myocardial fibrosis on cardiac function and clinical outcomes in patients with heart failure. In this regard, findings from the past 20 years suggest that various clinicopathological phenotypes of diffuse myocardial fibrosis exist in patients with heart failure. In this Review, we summarize the current knowledge on the mechanisms and detrimental consequences of diffuse myocardial fibrosis in heart failure. Furthermore, we discuss the validity and usefulness of available imaging techniques and circulating biomarkers to assess the clinicopathological variation in this lesion and to track its clinical evolution. Finally, we highlight the currently available and potential future therapeutic strategies aimed at personalizing the prevention and reversal of diffuse myocardial fibrosis in patients with heart failure.
Collapse
|
8
|
Techorueangwiwat C, Kanitsoraphan C, Hansrivijit P. Therapeutic implications of statins in heart failure with reduced ejection fraction and heart failure with preserved ejection fraction: a review of current literature. F1000Res 2021; 10:16. [PMID: 36873456 PMCID: PMC9982192 DOI: 10.12688/f1000research.28254.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2021] [Indexed: 11/20/2022] Open
Abstract
Statins are one of the standard treatments to prevent cardiovascular events such as coronary artery disease and heart failure (HF). However, data on the use of statins to improve clinical outcomes in patients with established HF remains controversial. We summarized available clinical studies which investigated the effects of statins on clinical outcomes in patients with HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Statins possess many pleiotropic effects in addition to lipid-lowering properties that positively affect the pathophysiology of HF. In HFrEF, data from two large randomized placebo-controlled trials did not show benefits of statins on mortality of patients with HFrEF. However, more recent prospective cohort studies and meta-analyses have shown decreased risk of mortality as well as cardiovascular hospitalization with statins treatment. In HFpEF, most prospective and retrospective cohort studies as well as meta analyses have consistently reported positive effects of statins, including reducing mortality and improving other clinical outcomes. Current evidence also suggests better outcomes with lipophilic statins in patients with HF. In summary, statins might be effective in improving survival and other clinical outcomes in patients with HF, especially for patients with HFpEF. Lipophilic statins might also be more beneficial for HF patients. Based on current evidence, statins did not cause harm and should be continued in HF patients who are already taking the medication. Further randomized controlled trials are needed to clarify the benefits of statins in HF patients.
Collapse
|
9
|
Soga F, Tanaka H, Tatsumi K, Mochizuki Y, Sano H, Toki H, Matsumoto K, Shite J, Takaoka H, Doi T, Hirata KI. Impact of Dapagliflozin on the Left Ventricular Diastolic Function in Diabetic Patients with Heart Failure Complicating Cardiovascular Risk Factors. Intern Med 2021; 60:2367-2374. [PMID: 34334588 PMCID: PMC8381170 DOI: 10.2169/internalmedicine.6127-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Objective Our aim was to investigate the impact of the sodium glucose cotransporter type 2 (SGLT2) inhibitor on the left ventricular (LV) diastolic function in type 2 diabetes mellitus (T2DM) patients with chronic heart failure (HF) complicating cardiovascular risk factors. Methods We analyzed data from our previous prospective multicenter study, in which we investigated the effect of dapagliflozin on the LV diastolic function of T2DM patients with stable HF at five institutions in Japan. Patients who had been taking at least 1 antidiabetic drug other than SGLT2 inhibitors started treatment with dapagliflozin. Echocardiography was performed at baseline and six months after the administration of dapagliflozin. Cardiovascular risk factors other than T2DM were age, gender, hypertension, dyslipidemia, history of cardiovascular events and overweight. Results The LV diastolic function, defined as the ratio of the mitral inflow E to the mitral e' annular velocities (E/e'), significantly decreased from 9.3 to 8.5 by six months after the administration of dapagliflozin (p=0.020) as previously reported. A multivariate logistic regression analysis showed that dyslipidemia was the only independent determinant of improvement in the E/e' after the administration of dapagliflozin among cardiovascular risk factors. Furthermore, the relative change in the E/e' from baseline to six months after the administration of dapagliflozin for HF patients with preserved ejection fraction (HFpEF) and dyslipidemia was significantly larger than that for HFpEF patients without dyslipidemia (-15.2% vs. 29.6%, p=0.014), but no such finding was observed in non-HFpEF patients. Conclusion SGLT2 inhibitors may exert a more beneficial effect on the LV diastolic function for T2DM patients with stable HF, especially those with complicating dyslipidemia, than existing treatments.
Collapse
Affiliation(s)
- Fumitaka Soga
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Hidekazu Tanaka
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Kazuhiro Tatsumi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
- Tatsumi Clinic, Japan
| | - Yasuhide Mochizuki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
- Division of Cardiology, Osaka Saiseikai Nakatsu Hospital, Japan
| | - Hiroyuki Sano
- Division of Cardiology, Aijinkai Takatsuki Hospital, Japan
| | - Hiromi Toki
- Division of Cardiology, Kobe Red Cross Hospital, Japan
| | - Kensuke Matsumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Junya Shite
- Division of Cardiology, Osaka Saiseikai Nakatsu Hospital, Japan
| | | | - Tomofumi Doi
- Division of Cardiology, Kobe Red Cross Hospital, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| |
Collapse
|
10
|
Wang Y, Yang MX, Tu Q, Tao LY, Liu G, An H, Zhang H, Jin JL, Fan JS, Du YF, Zheng JG, Ren JY. Impact of Prior Ischemic Stroke on Outcomes in Patients With Heart Failure - A Propensity-Matched Study. Circ J 2020; 84:1797-1806. [PMID: 32893260 DOI: 10.1253/circj.cj-20-0210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Whether ischemic stroke per se, rather than older age or additional comorbidities, accounts for the adverse prognosis of heart failure (HF) is uncertain. The present study examineed the intrinsic association of ischemic stroke with outcomes in a propensity-matched cohort. METHODS AND RESULTS Of 1,351 patients hospitalized with HF, 388 (28.7%) had prior ischemic stroke. Using propensity score for prior ischemic stroke, estimated for each patient, a matched cohort of 379 pairs of HF patients with and without prior ischemic stroke, balanced on 32 baseline characteristics was assembled. At 30 days, prior ischemic stroke was associated with significantly higher risks of the combined endpoint of all-cause death or readmission (hazard ratio [HR]: 1.91; 95% confidence interval [CI]: 1.38 to 2.65; P<0.001), all-cause death (HR: 2.08; 95% CI: 1.28 to 3.38; P=0.003), all-cause readmission (HR: 2.67; 95% CI: 1.78 to 4.01; P<0.001), and HF readmission (HR: 2.11; 95% CI: 1.19 to 3.72; P=0.010). Prior ischemic stroke was associated with a significantly higher risk of all 4 outcomes at both 6 months and 1 year. CONCLUSIONS Prior ischemic stroke was a potent and persistent risk predictor of death and readmission among patients with HF after accounting for clinical characteristics.
Collapse
Affiliation(s)
- Yu Wang
- Department of Cardiology, China-Japan Friendship Hospital
| | - Meng-Xi Yang
- Department of Cardiology, China-Japan Friendship Hospital
| | - Qiang Tu
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences
- University of Chinese Academy of Sciences
| | - Li-Yuan Tao
- Research Center of Clinical Epidemiology, Peking University Third Hospital
| | - Gang Liu
- Department of Cardiovascular Surgery, Peking University People's Hospital
| | - Hui An
- Department of Cardiology, Hebei General Hospital
| | - Hu Zhang
- Department of Cardiology, China-Japan Friendship Hospital
| | - Jiang-Li Jin
- Department of Neurology, China-Japan Friendship Hospital
| | - Jia-Sai Fan
- Department of Cardiology, China-Japan Friendship Hospital
| | - Yi-Fei Du
- Department of Cardiology, China-Japan Friendship Hospital
| | - Jin-Gang Zheng
- Department of Cardiology, China-Japan Friendship Hospital
| | - Jing-Yi Ren
- Department of Cardiology, China-Japan Friendship Hospital
- Vascular Health Research Center of Peking University Health Science Center
| |
Collapse
|
11
|
Tsigkou V, Siasos G, Bletsa E, Panoilia ME, Papastavrou A, Kokosias G, Oikonomou E, Papageorgiou N, Zaromitidou M, Marinos G, Vavuranakis M, Stefanadis C, Papavassiliou AG, Tousoulis D. The Predictive Role for ST2 in Patients with Acute Coronary Syndromes and Heart Failure. Curr Med Chem 2020; 27:4479-4493. [PMID: 31622195 DOI: 10.2174/0929867326666191016121630] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/06/2019] [Accepted: 10/04/2019] [Indexed: 12/17/2022]
Abstract
Intensive research has shed light on the utilization of novel biomarkers which facilitate the diagnosis and prognosis of patients with different medical problems. One of the most important biomarkers especially in the spectrum of heart failure is soluble ST2 (sST2: soluble Suppression of Tumorigenicity 2), which is involved in inflammation, fibrosis and cardiac stress. In the revised 2017 ACC/AHA/HFSA, "Focused Update Guidelines for the Management of Heart Failure" ST2 was given a class-IIa recommendation for the optimal risk assessment in patients with heart failure. Many studies indicate that not only baseline but also serial measurements of ST2 can accurately predict future cardiovascular events in patients with Acute Coronary Syndromes and heart failure. Therefore, in this review, we are going to discuss the studies about the prognostic significance of ST2 in patients with Acute Coronary Syndromes, acute and chronic heart failure.
Collapse
Affiliation(s)
- Vasiliki Tsigkou
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Gerasimos Siasos
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evanthia Bletsa
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria-Evi Panoilia
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Angeliki Papastavrou
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Kokosias
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nikolaos Papageorgiou
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marina Zaromitidou
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Marinos
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Athanasios G Papavassiliou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitris Tousoulis
- Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
12
|
Zhang Y, Wang YT, Koka S, Zhang Y, Hussain T, Li X. Simvastatin improves lysosome function via enhancing lysosome biogenesis in endothelial cells. Front Biosci (Landmark Ed) 2020; 25:283-298. [PMID: 31585890 DOI: 10.2741/4807] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nlrp3 inflammasomes were shown to play a critical role in triggering obesity-associated early onsets of cardiovascular complications such as endothelial barrier dysfunction with endothelial hyperpermeability. Statins prevent endothelial dysfunction and decrease cardiovascular risk in patients with obesity and diabetes. However, it remains unclear whether statin treatment for obesity-induced endothelial barrier dysfunction is in part due to the blockade of Nlrp3 inflammasome signaling axis. The results showed that simvastatin, a clinically and widely used statin, prevented free fatty acid-induced endothelial hyperpermeability and disruption of ZO-1 and VE-cadherin junctions in mouse microvascular endothelial cells (MVECs). This protective effect of simvastatin was largely due to improved lysosome function that attenuated lysosome injury-mediated Nlrp3 inflammasome activation and subsequent release of high mobility group box protein-1 (HMGB1). Mechanistically, simvastatin induces autophagy that promotes removal of damaged lysosomes and also promotes lysosome regeneration that preserves lysosome function. Collectively, simvastatin treatment improves lysosome function via enhancing lysosome biogenesis and its autophagic turnover, which may be an important mechanism to suppress Nlrp3 inflammasome activation and prevents endothelial hyperpermeability in obesity.
Collapse
Affiliation(s)
- Youzhi Zhang
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Yun-Ting Wang
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204
| | - Saisudha Koka
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204
| | - Yang Zhang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298
| | - Tahir Hussain
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204
| | - Xiang Li
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204,
| |
Collapse
|
13
|
Bielecka-Dabrowa A, Bytyçi I, Von Haehling S, Anker S, Jozwiak J, Rysz J, Hernandez AV, Bajraktari G, Mikhailidis DP, Banach M. Association of statin use and clinical outcomes in heart failure patients: a systematic review and meta-analysis. Lipids Health Dis 2019; 18:188. [PMID: 31672151 PMCID: PMC6822388 DOI: 10.1186/s12944-019-1135-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/16/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The role of statins in patients with heart failure (HF) of different levels of left ventricular ejection fraction (LVEF) remains unclear especially in the light of the absence of prospective data from randomized controlled trials (RCTs) in non-ischemic HF, and taking into account potential statins' prosarcopenic effects. We assessed the association of statin use with clinical outcomes in patients with HF. METHODS We searched PubMed, EMBASE, Scopus, Google Scholar and Cochrane Central until August 2018 for RCTs and prospective cohorts comparing clinical outcomes with statin vs non-statin use in patients with HF at different LVEF levels. We followed the guidelines of the 2009 PRISMA statement for reporting and applied independent extraction by multiple observers. Meta-analyses of hazard ratios (HRs) of effects of statins on clinical outcomes used generic inverse variance method and random model effects. Clinical outcomes were all-cause mortality, cardiovascular (CV) mortality and CV hospitalization. RESULTS Finally we included 17 studies (n = 88,100; 2 RCTs and 15 cohorts) comparing statin vs non-statin users (mean follow-up 36 months). Compared with non-statin use, statin use was associated with lower risk of all-cause mortality (HR 0.77, 95% confidence interval [CI], 0.72-0.83, P < 0.0001, I2 = 63%), CV mortality (HR 0.82, 95% CI: 0.76-0.88, P < 0.0001, I2 = 63%), and CV hospitalization (HR 0.78, 95% CI: 0.69-0.89, P = 0.0003, I2 = 36%). All-cause mortality was reduced on statin therapy in HF with both EF < 40% and ≥ 40% (HR: 0.77, 95% Cl: 0.68-0.86, P < 0.00001, and HR 0.75, 95% CI: 0.69-0.82, P < 0.00001, respectively). Similarly, CV mortality (HR 0.86, 95% CI: 0.79-0.93, P = 0.0003, and HR 0.83, 95% CI: 0.77-0.90, P < 0.00001, respectively), and CV hospitalizations (HR 0.80 95% CI: 0.64-0.99, P = 0.04 and HR 0.76 95% CI: 0.61-0.93, P = 0.009, respectively) were reduced in these EF subgroups. Significant effects on all clinical outcomes were also found in cohort studies' analyses; the effect was also larger and significant for lipophilic than hydrophilic statins. CONCLUSIONS In conclusion, statins may have a beneficial effect on CV outcomes irrespective of HF etiology and LVEF level. Lipophilic statins seem to be much more favorable for patients with heart failure.
Collapse
Affiliation(s)
- Agata Bielecka-Dabrowa
- Department of Hypertension, Medical University of Lodz, Rzgowska, 281/289; 93-338, Łódź, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Ibadete Bytyçi
- Clinic of Cardiology, University Clinical Centre of Kosovo, Prishtina, Republic of Kosovo
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Stephan Von Haehling
- Department of Cardiology and Pneumology, University Medical Center Gottingen (UMG), Gottingen, Germany
| | - Stefan Anker
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jacek Jozwiak
- Department of Family Medicine and Public Health, Institute of Medicine, University of Opole, Opole, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Lodz, Poland
| | - Adrian V Hernandez
- Health Outcomes, Policy, and Evidence Synthesis (HOPES) Group, University of Connecticut School of Pharmacy, Storrs, CT, USA
- School of Medicine, Universidad Peruana de Ciencias Aplicadas (UPC), Lima, Peru
| | - Gani Bajraktari
- Clinic of Cardiology, University Clinical Centre of Kosovo, Prishtina, Republic of Kosovo
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, UK
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Rzgowska, 281/289; 93-338, Łódź, Poland.
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| |
Collapse
|
14
|
Godoy JC, Niesman IR, Busija AR, Kassan A, Schilling JM, Schwarz A, Alvarez EA, Dalton ND, Drummond JC, Roth DM, Kararigas G, Patel HH, Zemljic-Harpf AE. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes. FASEB J 2018; 33:1209-1225. [PMID: 30169110 DOI: 10.1096/fj.201800876r] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Statins, which reduce LDL-cholesterol by inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, are among the most widely prescribed drugs. Skeletal myopathy is a known statin-induced adverse effect associated with mitochondrial changes. We hypothesized that similar effects would occur in cardiac myocytes in a lipophilicity-dependent manner between 2 common statins: atorvastatin (lipophilic) and pravastatin (hydrophilic). Neonatal cardiac ventricular myocytes were treated with atorvastatin and pravastatin for 48 h. Both statins induced endoplasmic reticular (ER) stress, but only atorvastatin inhibited ERK1/2T202/Y204, AktSer473, and mammalian target of rapamycin signaling; reduced protein abundance of caveolin-1, dystrophin, epidermal growth factor receptor, and insulin receptor-β; decreased Ras homolog gene family member A activation; and induced apoptosis. In cardiomyocyte-equivalent HL-1 cells, atorvastatin, but not pravastatin, reduced mitochondrial oxygen consumption. When male mice underwent atorvastatin and pravastatin administration per os for up to 7 mo, only long-term atorvastatin, but not pravastatin, induced elevated serum creatine kinase; swollen, misaligned, size-variable, and disconnected cardiac mitochondria; alteration of ER structure; repression of mitochondria- and endoplasmic reticulum-related genes; and a 21% increase in mortality in cardiac-specific vinculin-knockout mice during the first 2 months of administration. To our knowledge, we are the first to demonstrate in vivo that long-term atorvastatin administration alters cardiac ultrastructure, a finding with important clinical implications.-Godoy, J. C., Niesman, I. R., Busija, A. R., Kassan, A., Schilling, J. M., Schwarz, A., Alvarez, E. A., Dalton, N. D., Drummond, J. C., Roth, D. M., Kararigas, G., Patel, H. H., Zemljic-Harpf, A. E. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes.
Collapse
Affiliation(s)
- Joseph C Godoy
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Ingrid R Niesman
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna R Busija
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Adam Kassan
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, North Hollywood, California, USA
| | - Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna Schwarz
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Erika A Alvarez
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Nancy D Dalton
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - John C Drummond
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Georgios Kararigas
- Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Alice E Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
15
|
Bielecka-Dabrowa A, Fabis J, Mikhailidis DP, von Haehling S, Sahebkar A, Rysz J, Banach M. Prosarcopenic Effects of Statins May Limit Their Effectiveness in Patients with Heart Failure. Trends Pharmacol Sci 2018; 39:331-353. [DOI: 10.1016/j.tips.2018.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/25/2022]
|
16
|
Lin L, Xu C, Carraway MS, Piantadosi CA, Whorton AR, Li S. RhoA inactivation by S-nitrosylation regulates vascular smooth muscle contractive signaling. Nitric Oxide 2018; 74:56-64. [DOI: 10.1016/j.niox.2018.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 01/13/2018] [Accepted: 01/16/2018] [Indexed: 01/19/2023]
|
17
|
Oikawa T, Sakata Y, Nochioka K, Miura M, Tsuji K, Onose T, Abe R, Kasahara S, Sato M, Shiroto T, Takahashi J, Miyata S, Shimokawa H. Prognostic Impact of Statin Intensity in Heart Failure Patients With Ischemic Heart Disease: A Report From the CHART-2 (Chronic Heart Failure Registry and Analysis in the Tohoku District 2) Study. J Am Heart Assoc 2018. [PMID: 29540427 PMCID: PMC5907545 DOI: 10.1161/jaha.117.007524] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background The beneficial prognostic impact of statins has been established in patients with ischemic heart disease but not in those with heart failure (HF). In addition, it is still unclear whether patients benefit from statins regardless of low‐density lipoprotein cholesterol levels. Methods and Results We examined 2444 consecutive stage C or D HF patients with ischemic heart disease registered in CHART‐2 (Chronic Heart Failure Registry and Analysis in the Tohoku District 2), a multicenter, prospective, observational cohort study in Japan. Patients were divided into 3 groups according to the Japanese standard doses of statins and statin‐intensity categories defined by the 2013 American College of Cardiology and American Heart Association guidelines: higher (moderate‐high)‐intensity (n=868), lower (low)‐intensity (n=526), and no statin (n=1050). The median follow‐up period was 6.4 years (13929 person‐years). Analysis with the inverse probability of treatment weighted using a propensity score for multiple treatment revealed that both the higher‐intesity group (hazard ratio [HR]: 0.68; P<0.001) and the lower‐intensity group (HR: 0.82; P<0.001) had significantly lower incidence of the primary end point—a composite of all‐cause death and HF admission—compared with the no statin group. The higher‐intensity statin group had significantly lower incidence of the primary end point (HR: 0.82; P<0.001), all‐cause death (HR: 0.83; P<0.001), and HF admission (HR: 0.78; P<0.001) than the lower‐intensity statin group. Moreover, the use of statins, either higher‐ or lower‐intensity, was associated with reduced incidence of the primary end point, regardless of low‐density lipoprotein cholesterol levels. Conclusions These results suggest that statin use, particularly the use of higher‐intensity statins, has a beneficial prognostic impact in HF patients with ischemic heart disease, regardless of low‐density lipoprotein cholesterol levels. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT00418041.
Collapse
Affiliation(s)
- Takuya Oikawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiko Sakata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kotaro Nochioka
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanobu Miura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Evidence-Based Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kanako Tsuji
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeo Onose
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ruri Abe
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shintaro Kasahara
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Sato
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Shiroto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Miyata
- Department of Evidence-Based Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Evidence-Based Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | |
Collapse
|
18
|
Srivaratharajah K, Coutinho T, deKemp R, Liu P, Haddad H, Stadnick E, Davies RA, Chih S, Dwivedi G, Guo A, Wells GA, Bernick J, Beanlands R, Mielniczuk LM. Reduced Myocardial Flow in Heart Failure Patients With Preserved Ejection Fraction. Circ Heart Fail 2017; 9:CIRCHEARTFAILURE.115.002562. [PMID: 27413034 DOI: 10.1161/circheartfailure.115.002562] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 05/12/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND There remains limited insight into the pathophysiology and therapeutic advances directed at improving prognosis for patients with heart failure with preserved ejection fraction (HFpEF). Recent studies have suggested a role for coronary microvascular dysfunction in HFpEF. Rb-82 cardiac positron emission tomography imaging is a noninvasive, quantitative approach to measuring myocardial flow reserve (MFR), a surrogate marker for coronary vascular health. The aim of this study was to determine whether abnormalities exist in MFR in patients with HFpEF without epicardial coronary artery disease. METHODS AND RESULTS A total of 376 patients with ejection fraction ≥50%, no known history of obstructive coronary artery disease, and a confirmed diagnosis of heart failure (n=78) were compared with patients with no evidence of heart failure (n=298), further stratified into those with (n=186) and without (n=112) hypertension. Global and regional left ventricular MFR was calculated as stress/rest myocardial blood flow using Rb-82 positron emission tomography. Patients with HFpEF were more likely to be older, female, and have comorbid hypertension, diabetes mellitus, dyslipidemia, atrial fibrillation, anemia, and renal dysfunction. HFpEF was associated with a significant reduction in global MFR (2.16±0.69 in HFpEF versus 2.54±0.80 in hypertensive controls; P<0.02 and 2.89±0.70 in normotensive controls; P<0.001). A diagnosis of HFpEF was associated with 2.62 times greater unadjusted odds of having low global MFR (defined as <2.0) and remained a significant predictor of reduced global MFR after adjusting for comorbidities. CONCLUSIONS HFpEF, in the absence of known history for obstructive epicardial coronary artery disease, is associated with reduced MFR independent of other risk factors.
Collapse
Affiliation(s)
| | - Thais Coutinho
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Robert deKemp
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Peter Liu
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Haissam Haddad
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Ellamae Stadnick
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Ross A Davies
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Sharon Chih
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Girish Dwivedi
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Ann Guo
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - George A Wells
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Jordan Bernick
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Robert Beanlands
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada
| | - Lisa M Mielniczuk
- From the Division of Cardiology, University of Ottawa Heart Institute, Ontario, Canada.
| |
Collapse
|
19
|
Statins and oxidative stress in chronic heart failure. Rev Port Cardiol 2016; 35:41-57. [PMID: 26763895 DOI: 10.1016/j.repc.2015.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/13/2015] [Indexed: 11/23/2022] Open
Abstract
Statins are the most commonly prescribed drugs for the treatment of dyslipidemia. They are also recommended in primary and secondary prevention of cardiovascular disease. In addition to decreasing cholesterol synthesis, statins interfere with the synthesis of isoprenoid intermediates, which may explain many of their pleiotropic properties, including their antioxidant effects. Oxidative stress is defined as an imbalance between the synthesis of reactive oxygen species and their elimination by antioxidant defense systems, with a prevailing pro-oxidant status that results in macromolecular damage and disruption of cellular redox signaling. Reactive oxygen species interfere with various processes that affect cardiac structure and function, contributing to the contractile dysfunction, myocardial hypertrophy and fibrosis observed in the pathophysiology of heart failure. By regulating several molecular pathways that control nicotinamide adenine dinucleotide phosphate oxidase and endothelial nitric oxide synthase activity, statins help restore redox homeostasis. These drugs also contribute to the control of inflammation and appear to have a protective role in various diseases. The results of observational studies and clinical trials with statins in heart failure have not been consensual. This review aims to analyze the role of oxidative stress in heart failure and the molecular mechanisms underlying statins' antioxidant properties. It also examines current scientific evidence on the use of these drugs as a specific treatment for heart failure.
Collapse
|
20
|
Statins and oxidative stress in chronic heart failure. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2016. [DOI: 10.1016/j.repce.2015.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
21
|
Affiliation(s)
- Anping Cai
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China (A.C., Y.Z., L.L.)
| | - Yingling Zhou
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China (A.C., Y.Z., L.L.)
| | - Liwen Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China (A.C., Y.Z., L.L.)
| |
Collapse
|
22
|
Gutiérrez-Vargas JA, Cespedes-Rubio A, Cardona-Gómez GP. Perspective of synaptic protection after post-infarction treatment with statins. J Transl Med 2015; 13:118. [PMID: 25884826 PMCID: PMC4403706 DOI: 10.1186/s12967-015-0472-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/23/2015] [Indexed: 11/30/2022] Open
Abstract
Stroke is the second most common cause of death in people over 45 years of age in Colombia and is the leading cause of permanent disability worldwide. Cerebral ischemia is a stroke characterized by decreased blood flow due to the occlusion of one or more cerebral arteries, which can cause memory problems and hemiplegia or paralysis, among other impairments. The literature contains hundreds of therapies (invasive and noninvasive) that exhibit a neuroprotective effect when evaluated in animal models. However, in clinical trials, most of these drugs do not reproduce the previously demonstrated neuroprotective property, and some even have adverse effects that had not previously been detected in animal experimentation. Statins are drugs that inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, the rate-limiting enzyme in cholesterol synthesis. Several studies have shown that statin therapy in an animal model of focal cerebral ischemia reduces infarct volume, as well as markers of neurodegeneration, activates neuronal survival pathways, and improves performance on learning and memory tests. Given the implied therapeutic benefit and the limited understanding of the mechanism of action of statins in brain repair, it is necessary to address the biochemical and tissue effects of these drugs on synaptic proteins, such as NMDA receptors, synaptic adhesion proteins, and cytoskeletal proteins; these proteins are highly relevant therapeutic targets, which, in addition to giving a structural account of synaptic connectivity and function, are also indicators of cellular communication and the integrity of the blood–brain barrier, which are widely affected in the long term post-cerebral infarct but, interestingly, are protected by statins when administered during the acute phase.
Collapse
Affiliation(s)
- Johanna Andrea Gutiérrez-Vargas
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, School of Medicine, SIU, University of Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Angel Cespedes-Rubio
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, School of Medicine, SIU, University of Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia. .,Neurodegenerative Diseases Research Group, Department of Animal Health - Faculty of Veterinary Medicine - University of Tolima, Ibague, Colombia.
| | - Gloria Patricia Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, School of Medicine, SIU, University of Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
23
|
Mozos I. Arrhythmia risk in liver cirrhosis. World J Hepatol 2015; 7:662-672. [PMID: 25866603 PMCID: PMC4388994 DOI: 10.4254/wjh.v7.i4.662] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 12/04/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
Interactions between the functioning of the heart and the liver have been described, with heart diseases affecting the liver, liver diseases affecting the heart, and conditions that simultaneously affect both. The heart is one of the most adversely affected organs in patients with liver cirrhosis. For example, arrhythmias and electrocardiographic changes are observed in patients with liver cirrhosis. The risk for arrhythmia is influenced by factors such as cirrhotic cardiomyopathy, cardiac ion channel remodeling, electrolyte imbalances, impaired autonomic function, hepatorenal syndrome, metabolic abnormalities, advanced age, inflammatory syndrome, stressful events, impaired drug metabolism and comorbidities. Close monitoring of cirrhotic patients is needed for arrhythmias, particularly when QT interval-prolonging drugs are given, or if electrolyte imbalances or hepatorenal syndrome appear. Arrhythmia risk may persist after liver transplantation due to possible QT interval prolongation, persistence of the parasympathetic impairment, post-transplant reperfusion and chronic immunosuppression, as well as consideration of the fact that the transplant itself is a stressful event for the cardiovascular system. The aims of the present article were to provide a review of the most important data regarding the epidemiology, pathophysiology, and biomarkers of arrhythmia risk in patients with liver cirrhosis, to elucidate the association with long-term outcome, and to propose future research directions.
Collapse
|
24
|
Oikonomou E, Siasos G, Zaromitidou M, Hatzis G, Mourouzis K, Chrysohoou C, Zisimos K, Mazaris S, Tourikis P, Athanasiou D, Stefanadis C, Papavassiliou AG, Tousoulis D. Atorvastatin treatment improves endothelial function through endothelial progenitor cells mobilization in ischemic heart failure patients. Atherosclerosis 2015; 238:159-164. [PMID: 25525743 DOI: 10.1016/j.atherosclerosis.2014.12.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/19/2014] [Accepted: 12/01/2014] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Endothelial function is an independent predictor of prognosis in heart failure (HF) subjects. Statins, beyond their lipid lowering role, exert beneficial effect in patients with atherosclerosis. In the present study we examined the impact of low and intermediate dose atorvastatin treatment on endothelial function, bone marrow-derived endothelial progenitor cells (EPC) mobilization and inflammatory status according to HF patient status. METHODS We studied the effect of 4 weeks administration of atorvastatin in 26 patients with ischemic HF. The study was carried out on two separate arms, one with atorvastatin 40 mg/d and one with atorvastatin 10 mg/d (randomized, double-blind, cross-over design). The number of circulating CD34(+)/CD133(+)/KDR(+) EPCs was evaluated by flow cytometry. Endothelial function was evaluated by flow mediated dilation (FMD) in the brachial artery. Serum levels of tumor necrosis factor alpha (TNF-α) were measured by ELISA. RESULTS Treatment with atorvastatin 40 mg/d significantly increased circulating EPC (p = 0.002), FMD (p = 0.001) and reduced TNF-α (p = 0.01) compared to baseline. Similarly, treatment with atorvastatin 10 mg/day increased circulating EPC (p = 0.01), FMD (p = 0.08) and reduced TNF-α (p = 0.01) compared to baseline. Interestingly, with 40 mg/day atorvastatin treatment the increase in EPC was higher in subjects categorized as NYHA class II compared to subjects categorized as NYHA class III (p = 0.03). CONCLUSIONS Our results confirmed the distinct impact of atorvastatin treatment on the restoration of endothelial function due to EPC mobilization in ischemic HF subjects. Moreover, these findings provide the potential clinical significance of EPC status monitoring to individualize treatment in HF subjects.
Collapse
Affiliation(s)
- Evangelos Oikonomou
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Gerasimos Siasos
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece; Department of Biological Chemistry, University of Athens Medical School, Athens, Greece.
| | - Marina Zaromitidou
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - George Hatzis
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Konstantinos Mourouzis
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Christine Chrysohoou
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Konstantinos Zisimos
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Savvas Mazaris
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Panagiotis Tourikis
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Dimitris Athanasiou
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Christodoulos Stefanadis
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Dimitris Tousoulis
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| |
Collapse
|
25
|
Morimoto T, Katanasaka Y, Sunagawa Y, Hirano S, Miyazaki Y, Funamoto M, Hojo Y, Suzuki H, Morimoto E, Ueno M, Shimatsu A, Satoh-Asahara N, Yamakage H, Wada H, Hasegawa K. Effects of Statins on Left Ventricular Diastolic Function in Patients with Dyslipidemia and Diastolic Dysfunction (Stat-LVDF Study). Biol Pharm Bull 2015; 38:1404-9. [DOI: 10.1248/bpb.b15-00126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Shizuoka General Hospital
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Yasufumi Katanasaka
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Shizuoka General Hospital
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Shizuoka General Hospital
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Sae Hirano
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Shizuoka Saiseikai General Hospital
| | - Yusuke Miyazaki
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | - Masafumi Funamoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | - Yuya Hojo
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | - Hidetoshi Suzuki
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine
| | - Akira Shimatsu
- Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Noriko Satoh-Asahara
- Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Hajime Yamakage
- Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Hiromichi Wada
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Koji Hasegawa
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| |
Collapse
|
26
|
Tousoulis D, Oikonomou E, Siasos G, Stefanadis C. Diabetes Mellitus and Heart Failure. Eur Cardiol 2014; 9:37-42. [PMID: 30310483 PMCID: PMC6159396 DOI: 10.15420/ecr.2014.9.1.37] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/13/2014] [Indexed: 12/24/2022] Open
Abstract
Diabetes mellitus and heart failure are two multifaceted entities characterised by high morbidity and mortality. Early epidemiological and prospective studies have observed the frequent co-existence of both conditions. Importantly, diabetes mellitus can precipitate or worsen heart failure due to the accumulation of advanced glycation end products, oxidative stress, inflammatory status impairment, decay of intracellular calcium, changes in microRNAs expression, not to mention atherosclerosis progression and coronary artery disease. Heart failure also impairs glucose metabolism through less well-known mechanisms. Attention must especially be given in the treatment as there are frequently adverse interactions between the two diseases and novel agents against diabetic cardiomyopathy are under investigation. As several missing links still exist in the connection between heart failure and diabetes mellitus we will review, in this article, the most recent data underlying the interaction of them and provide an overview of the most important clinical perspectives.
Collapse
Affiliation(s)
- Dimitris Tousoulis
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Evangelos Oikonomou
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Gerasimos Siasos
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| | - Christodoulos Stefanadis
- 1st Cardiology Department, University of Athens Medical School, "Hippokration" Hospital, Athens, Greece
| |
Collapse
|
27
|
Du G, Song Y, Zhang T, Ma L, Bian N, Chen X, Feng J, Chang Q, Li Z. Simvastatin attenuates TNF‑α‑induced apoptosis in endothelial progenitor cells via the upregulation of SIRT1. Int J Mol Med 2014; 34:177-82. [PMID: 24718722 DOI: 10.3892/ijmm.2014.1740] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/01/2014] [Indexed: 01/30/2023] Open
Abstract
Endothelial progenitor cells (EPCs) originate from the bone marrow and can be classified as either early or late EPCs. The focus of this study was on late EPCs, as they play an important role in angiogenesis and vascular proliferation. Evidence suggests that inflammatory and oxidative changes can increase EPC apoptosis. Of note, tumor necrosis factor-α (TNF-α) is a contributing risk factor to the development of atherosclerosis and plays a key role as both an inflammatory mediator and an inducer of apoptosis in endothelial cells. Additionally, a member of the sirtuin family, silent information regulator type-1 (SIRT1), promotes cell survival by repressing p53- and non-p53-dependent apoptosis in response to DNA damage and oxidative stress. Statins have also been shown to play a key role in the prevention of endothelial apoptosis and senescence via their lipid-lowering and anti-inflammatory actions. However, there is little evidence that statins themselves attenuate EPC apoptosis induced by TNF-α. The aim of this study was to demonstrate the effectiveness of one of the most commonly used statins, simvastatin, on decreasing TNF-α-induced apoptosis in EPCs. The results indicated that SIRT1 protein expression was decreased by TNF-α in a time- and dose-dependent manner and that while TNF-α caused a marked increase in the percentage of apoptotic EPCs, application of simvastatin decreased this percentage. A high concentration of simvastatin promoted the expression of SIRT1 and increased the proliferation of EPCs. In conclusion, findings of this study showed that simvastatin is crucial in counteracting the TNF-α-induced apoptosis of EPCs and that this protection may involve the actions of SIRT1.
Collapse
Affiliation(s)
- Gang Du
- Department of Internal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Yunlin Song
- Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, P.R. China
| | - Tao Zhang
- Department of Internal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Long Ma
- Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, P.R. China
| | - Ning Bian
- Department of Internal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Xiaoming Chen
- Department of Internal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Jianyi Feng
- Department of Internal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Qing Chang
- Department of Histology and Embryology, Medical College of Jinan University, Guangzhou 510632, P.R. China
| | - Zicheng Li
- Department of Internal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| |
Collapse
|