1
|
Dong Q, Dai G, Quan N, Tong Q. Role of natural products in cardiovascular disease. Mol Cell Biochem 2024. [DOI: 10.1007/s11010-024-05048-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/09/2024] [Indexed: 01/03/2025]
|
2
|
Zhang M, Wei J, Sun Y, He C, Ma S, Pan X, Zhu X. The efferocytosis process in aging: Supporting evidence, mechanisms, and therapeutic prospects for age-related diseases. J Adv Res 2024:S2090-1232(24)00109-7. [PMID: 38499245 DOI: 10.1016/j.jare.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Aging is characterized by an ongoing struggle between the buildup of damage caused by a combination of external and internal factors. Aging has different effects on phagocytes, including impaired efferocytosis. A deficiency in efferocytosis can cause chronic inflammation, aging, and several other clinical disorders. AIM OF REVIEW Our review underscores the possible feasibility and extensive scope of employing dual targets in various age-related diseases to reduce the occurrence and progression of age-related diseases, ultimately fostering healthy aging and increasing lifespan. Key scientific concepts of review Hence, the concurrent implementation of strategies aimed at augmenting efferocytic mechanisms and anti-aging treatments has the potential to serve as a potent intervention for extending the duration of a healthy lifespan. In this review, we comprehensively discuss the concept and physiological effects of efferocytosis. Subsequently, we investigated the association between efferocytosis and the hallmarks of aging. Finally, we discuss growing evidence regarding therapeutic interventions for age-related disorders, focusing on the physiological processes of aging and efferocytosis.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
3
|
Rolski F, Tkacz K, Węglarczyk K, Kwiatkowski G, Pelczar P, Jaźwa-Kusior A, Bar A, Kuster GM, Chłopicki S, Siedlar M, Kania G, Błyszczuk P. TNF-α protects from exacerbated myocarditis and cardiac death by suppressing expansion of activated heart-reactive CD4+ T cells. Cardiovasc Res 2024; 120:82-94. [PMID: 37879102 PMCID: PMC10898940 DOI: 10.1093/cvr/cvad158] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 08/08/2023] [Accepted: 09/26/2023] [Indexed: 10/27/2023] Open
Abstract
AIMS Tumour necrosis factor α (TNF-α) represents a classical pro-inflammatory cytokine, and its increased levels positively correlate with the severity of many cardiovascular diseases. Surprisingly, some heart failure patients receiving high doses of anti-TNF-α antibodies showed serious health worsening. This work aimed to examine the role of TNF-α signalling on the development and progression of myocarditis and heart-specific autoimmunity. METHODS AND RESULTS Mice with genetic deletion of TNF-α (Tnf+/- and Tnf-/-) and littermate controls (Tnf+/+) were used to study myocarditis in the inducible and the transgenic T cell receptor (TCRM) models. Tnf+/- and Tnf-/- mice immunized with α-myosin heavy chain peptide (αMyHC) showed reduced myocarditis incidence, but the susceptible animals developed extensive inflammation in the heart. In the TCRM model, defective TNF-α production was associated with increased mortality at a young age due to cardiomyopathy and cardiac fibrosis. We could confirm that TNF-α as well as the secretome of antigen-activated heart-reactive effector CD4+ T (Teff) cells effectively activated the adhesive properties of cardiac microvascular endothelial cells (cMVECs). Our data suggested that TNF-α produced by endothelial in addition to Teff cells promoted leucocyte adhesion to activated cMVECs. Analysis of CD4+ T lymphocytes from both models of myocarditis showed a strongly increased fraction of Teff cells in hearts, spleens, and in the blood of Tnf+/- and Tnf-/- mice. Indeed, antigen-activated Tnf-/- Teff cells showed prolonged long-term survival and TNF-α cytokine-induced cell death of heart-reactive Teff. CONCLUSION TNF-α signalling promotes myocarditis development by activating cardiac endothelial cells. However, in the case of established disease, TNF-α protects from exacerbating cardiac inflammation by inducing activation-induced cell death of heart-reactive Teff. These data might explain the lack of success of standard anti-TNF-α therapy in heart failure patients and open perspectives for T cell-targeted approaches.
Collapse
Affiliation(s)
- Filip Rolski
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, Cracow 30-663, Poland
| | - Karolina Tkacz
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, Cracow 30-663, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, Cracow 30-663, Poland
| | - Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Cracow, Poland
| | - Paweł Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | | | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Cracow, Poland
| | - Gabriela M Kuster
- Clinic of Cardiology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Stefan Chłopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Cracow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, Cracow 30-663, Poland
| | - Gabriela Kania
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Przemysław Błyszczuk
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, Cracow 30-663, Poland
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| |
Collapse
|
4
|
Ang SP, Chia JE, Jaiswal V, Hanif M, Iglesias J. Prognostic Value of Neutrophil-to-Lymphocyte Ratio in Patients with Acute Decompensated Heart Failure: A Meta-Analysis. J Clin Med 2024; 13:1212. [PMID: 38592030 PMCID: PMC10931846 DOI: 10.3390/jcm13051212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Inflammation plays a pivotal role in the pathogenesis of both acute and chronic heart failure. Recent studies showed that the neutrophil-to-lymphocyte ratio (NLR) could be related to adverse outcomes in patients with cardiovascular diseases. We sought to evaluate whether NLR could predict mortality in patients with acute heart failure by means of a meta-analysis. Methods: A comprehensive literature search was performed in PubMed, Embase, and Cochrane databases through January 2023 for studies evaluating the association of NLR with mortality in patients with acute heart failure. Primary outcomes were in-hospital mortality and long-term all-cause mortality. Endpoints were pooled using a random-effects DerSimonian-and-Laird model and were expressed as a hazard ratio (HR) or mean difference (MD) with their corresponding 95% confidence intervals. Results: A total of 15 studies with 15,995 patients with acute heart failure were included in the final study. Stratifying patients based on a cut-off NLR, we found that high NLR was associated with a significantly higher in-hospital mortality [HR 1.54, 95% CI (1.18-2.00), p < 0.001] and long-term all-cause mortality [HR 1.61, 95% CI (1.40-1.86), p < 0.001] compared to the low-NLR group. Comparing the highest against the lowest NLR quartile, it was shown that patients in the highest NLR quartile has a significantly heightened risk of long-term all-cause mortality [HR 1.77, 95% CI (1.38-2.26), p < 0.001] compared to that of lowest NLR quartile. However, the risks of in-hospital mortality were compared between both quartiles of patients [HR 1.78, 95% CI (0.91-3.47), p = 0.09]. Lastly, NLR values were significantly elevated among non-survivors compared to survivors during index hospitalization [MD 5.07, 95% CI (3.34-6.80), p < 0.001] and during the follow-up period [MD 1.06, 95% CI (0.54-1.57), p < 0.001]. Conclusions: Elevated NLR was associated with an increased risk of short- and long-term mortality and could be a useful tool or incorporated in the risk stratification in patients with acute heart failure.
Collapse
Affiliation(s)
- Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ 08755, USA;
| | - Jia Ee Chia
- Department of Internal Medicine, Texas Tech University Health Science Center, El Paso, TX 79905, USA;
| | - Vikash Jaiswal
- Department of Internal Medicine, Larkin Community Hospital, South Miami, FL 33143, USA;
| | - Muhammad Hanif
- Department of Internal Medicine, Suny Upstate Medical University, Syracuse, NY 13210, USA;
| | - Jose Iglesias
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ 08755, USA;
- Department of Internal Medicine, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| |
Collapse
|
5
|
Bhullar SK, Dhalla NS. Adaptive and maladaptive roles of different angiotensin receptors in the development of cardiac hypertrophy and heart failure. Can J Physiol Pharmacol 2024; 102:86-104. [PMID: 37748204 DOI: 10.1139/cjpp-2023-0226] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Angiotensin II (Ang II) is formed by the action of angiotensin-converting enzyme (ACE) in the renin-angiotensin system. This hormone is known to induce cardiac hypertrophy and heart failure and its actions are mediated by the interaction of both pro- and antihypertrophic Ang II receptors (AT1R and AT2R). Ang II is also metabolized by ACE 2 to Ang-(1-7), which elicits the activation of Mas receptors (MasR) for inducing antihypertrophic actions. Since heart failure under different pathophysiological situations is preceded by adaptive and maladaptive cardiac hypertrophy, we have reviewed the existing literature to gain some information regarding the roles of AT1R, AT2R, and MasR in both acute and chronic conditions of cardiac hypertrophy. It appears that the activation of AT1R may be involved in the development of adaptive and maladaptive cardiac hypertrophy as well as subsequent heart failure because both ACE inhibitors and AT1R antagonists exert beneficial effects. On the other hand, the activation of both AT2R and MasR may prevent the occurrence of maladaptive cardiac hypertrophy and delay the progression of heart failure, and thus therapy with different activators of these antihypertrophic receptors under chronic pathological stages may prove beneficial. Accordingly, it is suggested that a great deal of effort should be made to develop appropriate activators of both AT2R and MasR for the treatment of heart failure subjects.
Collapse
Affiliation(s)
- Sukhwinder K Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
6
|
Dai G, Li M, Xu H, Quan N. Status of Research on Sestrin2 and Prospects for its Application in Therapeutic Strategies Targeting Myocardial Aging. Curr Probl Cardiol 2023; 48:101910. [PMID: 37422038 DOI: 10.1016/j.cpcardiol.2023.101910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
Cardiac aging is accompanied by changes in the heart at the cellular and molecular levels, leading to alterations in cardiac structure and function. Given today's increasingly aging population, the decline in cardiac function caused by cardiac aging has a significant impact on quality of life. Antiaging therapies to slow the aging process and attenuate changes in cardiac structure and function have become an important research topic. Treatment with drugs, including metformin, spermidine, rapamycin, resveratrol, astaxanthin, Huolisu oral liquid, and sulforaphane, has been demonstrated be effective in delaying cardiac aging by stimulating autophagy, delaying ventricular remodeling, and reducing oxidative stress and the inflammatory response. Furthermore, caloric restriction has been shown to play an important role in delaying aging of the heart. Many studies in cardiac aging and cardiac aging-related models have demonstrated that Sestrin2 has antioxidant and anti-inflammatory effects, stimulates autophagy, delays aging, regulates mitochondrial function, and inhibits myocardial remodeling by regulation of relevant signaling pathways. Therefore, Sestrin2 is likely to become an important target for antimyocardial aging therapy.
Collapse
Affiliation(s)
- Gaoying Dai
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Meina Li
- Department of Infection Control, The First Hospital of Jilin University, Changchun, China
| | - He Xu
- Department of Integrative Medicine, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Nanhu Quan
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
7
|
Myostatin deficiency decreases cardiac extracellular matrix in pigs. Transgenic Res 2022; 31:553-565. [PMID: 35978205 DOI: 10.1007/s11248-022-00322-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/03/2022] [Indexed: 10/15/2022]
Abstract
Myostatin (MSTN), a member of the TGF-β superfamily, negatively regulates muscle growth. MSTN inhibition has been known to cause a double-muscled phenotype in skeletal muscle and fibrosis reduction in the heart. However, the role of MSTN in the cardiac extracellular matrix (ECM) needs more studies in various species of animal models to draw more objective conclusions. The main objective of the present study was to investigate whether loss of MSTN affects the cardiac extracellular matrix in pigs. Three MSTN knockouts (MSTN-/-) and three wild type (WT) male pigs were generated by crossing MSTN ± heterozygous gilts and boars. Cardiac ECM and underlying mechanisms were determined post-mortem. The role of MSTN on collagen expression was investigated by treating cardiac fibroblasts with active MSTN protein in vitro. MSTN protein was detected in WT hearts, while no expression was detected in MSTN-/- hearts. The heart-to-body weight ratio was significantly decreased in MSTN-/- pigs. The morphometric analyses, including picrosirius red staining, immunofluorescent staining, and ultra-structural thickness examination of the endomysium, revealed a significant reduction of connective tissue content in MSTN-/- hearts compared to WT. Hydroxyproline, type I collagen (Col1A), and p-Smad3/Smad3 levels were significantly lower in MSTN-/- hearts in vivo. On the contrary, cardiac fibroblasts treated with exogenous MSTN protein overexpressed Col1A and activated Smad and AKT signaling pathways in vitro. The present study suggests that inhibition of MSTN decreases cardiac extracellular matrix.
Collapse
|
8
|
Schimmel K, Ichimura K, Reddy S, Haddad F, Spiekerkoetter E. Cardiac Fibrosis in the Pressure Overloaded Left and Right Ventricle as a Therapeutic Target. Front Cardiovasc Med 2022; 9:886553. [PMID: 35600469 PMCID: PMC9120363 DOI: 10.3389/fcvm.2022.886553] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022] Open
Abstract
Myocardial fibrosis is a remodeling process of the extracellular matrix (ECM) following cardiac stress. "Replacement fibrosis" is a term used to describe wound healing in the acute phase of an injury, such as myocardial infarction. In striking contrast, ECM remodeling following chronic pressure overload insidiously develops over time as "reactive fibrosis" leading to diffuse interstitial and perivascular collagen deposition that continuously perturbs the function of the left (L) or the right ventricle (RV). Examples for pressure-overload conditions resulting in reactive fibrosis in the LV are systemic hypertension or aortic stenosis, whereas pulmonary arterial hypertension (PAH) or congenital heart disease with right sided obstructive lesions such as pulmonary stenosis result in RV reactive fibrosis. In-depth phenotyping of cardiac fibrosis has made it increasingly clear that both forms, replacement and reactive fibrosis co-exist in various etiologies of heart failure. While the role of fibrosis in the pathogenesis of RV heart failure needs further assessment, reactive fibrosis in the LV is a pathological hallmark of adverse cardiac remodeling that is correlated with or potentially might even drive both development and progression of heart failure (HF). Further, LV reactive fibrosis predicts adverse outcome in various myocardial diseases and contributes to arrhythmias. The ability to effectively block pathological ECM remodeling of the LV is therefore an important medical need. At a cellular level, the cardiac fibroblast takes center stage in reactive fibrotic remodeling of the heart. Activation and proliferation of endogenous fibroblast populations are the major source of synthesis, secretion, and deposition of collagens in response to a variety of stimuli. Enzymes residing in the ECM are responsible for collagen maturation and cross-linking. Highly cross-linked type I collagen stiffens the ventricles and predominates over more elastic type III collagen in pressure-overloaded conditions. Research has attempted to identify pro-fibrotic drivers causing fibrotic remodeling. Single key factors such as Transforming Growth Factor β (TGFβ) have been described and subsequently targeted to test their usefulness in inhibiting fibrosis in cultured fibroblasts of the ventricles, and in animal models of cardiac fibrosis. More recently, modulation of phenotypic behaviors like inhibition of proliferating fibroblasts has emerged as a strategy to reduce pathogenic cardiac fibroblast numbers in the heart. Some studies targeting LV reactive fibrosis as outlined above have successfully led to improvements of cardiac structure and function in relevant animal models. For the RV, fibrosis research is needed to better understand the evolution and roles of fibrosis in RV failure. RV fibrosis is seen as an integral part of RV remodeling and presents at varying degrees in patients with PAH and animal models replicating the disease of RV afterload. The extent to which ECM remodeling impacts RV function and thus patient survival is less clear. In this review, we describe differences as well as common characteristics and key players in ECM remodeling of the LV vs. the RV in response to pressure overload. We review pre-clinical studies assessing the effect of anti-fibrotic drug candidates on LV and RV function and their premise for clinical testing. Finally, we discuss the mode of action, safety and efficacy of anti-fibrotic drugs currently tested for the treatment of left HF in clinical trials, which might guide development of new approaches to target right heart failure. We touch upon important considerations and knowledge gaps to be addressed for future clinical testing of anti-fibrotic cardiac therapies.
Collapse
Affiliation(s)
- Katharina Schimmel
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Kenzo Ichimura
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Sushma Reddy
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - Francois Haddad
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Cardiovascular Medicine, Stanford University, Stanford, CA, United States
| | - Edda Spiekerkoetter
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,*Correspondence: Edda Spiekerkoetter,
| |
Collapse
|
9
|
Feliciano RDS, Manchini MT, Atum ALB, da Silva GA, Antônio EL, Serra AJ, Tucci PJF, Andrade de Mello R, Chavantes MC, Baltatu OC, Silva Júnior JA. Photobiomodulation therapy's effects on cardiac fibrosis activation after experimental myocardial infarction. Lasers Surg Med 2022; 54:883-894. [PMID: 35366381 DOI: 10.1002/lsm.23544] [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: 07/26/2021] [Revised: 02/14/2022] [Accepted: 03/18/2022] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Ischemic heart disease is the leading cause of death worldwide, and interventions to reduce myocardial infarction (MI) complications are widely researched. Photobiomodulation therapy (PBMT) has altered multiple biological processes in tissues and organs, including the heart. OBJECTIVES This study aimed to assess the temporal effects of PBMT on cardiac fibrosis activation after MI in rats. In this proof-of-concept study, we monitored the change in expression patterns over time of genes and microRNAs (miRNAs) involved in the formation of cardiac fibrosis post-MI submitted to PBMT. MATERIALS AND METHODS Experimental MI was induced, and PBMT was applied shortly after coronary artery ligation (laser light of wavelength 660 nm, 15 mW of power, energy density 22.5 J/cm2 , 60 seconds of application, irradiated area 0.785 cm2 , fluence 1.1 J/cm2 ). Ventricular septal samples were collected at 30 minutes, 3, 6, 24 hours, and 3 days post-MI to determine temporal PBMT's effects on messenger RNA (mRNA) expression associated with cardiac fibrosis activation and miRNAs expression. RESULTS PBMT, when applied after ischemia, reversed the changes in mRNA expression of myocardial extracellular matrix genes induced by MI. Surprisingly, PBMT modified cardiac miRNAs expression related to fibrosis replacement in the myocardium. Expression correlations between myocardial mRNAs were assessed. The correlation coefficient between miRNAs and target mRNAs was also determined. A positive correlation was detected among miR-21 and transforming growth factor beta-1 mRNA. The miR-29a expression negatively correlated to Col1a1, Col3a1, and MMP-2 mRNA expressions. In addition, we observed that miR-133 and Col1a1 mRNA were negatively correlated. CONCLUSION The results suggest that PBMT, through the modulation of gene transcription and miRNA expressions, can interfere in cardiac fibrosis activation after MI, mainly reversing the signaling pathway of profibrotic genes.
Collapse
Affiliation(s)
| | - Martha T Manchini
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil.,Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Allan L B Atum
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | | | - Ednei L Antônio
- Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Andrey J Serra
- Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paulo J F Tucci
- Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ramon Andrade de Mello
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - Maria C Chavantes
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - Ovidiu C Baltatu
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.,Center of Innovation, Technology and Education (CITE), Anhembi Morumbi University-Anima Institute, São José dos Campos, Brazil
| | - José A Silva Júnior
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| |
Collapse
|
10
|
Martens CR, Dorn LE, Kenney AD, Bansal SS, Yount JS, Accornero F. BEX1 is a critical determinant of viral myocarditis. PLoS Pathog 2022; 18:e1010342. [PMID: 35192678 PMCID: PMC8896894 DOI: 10.1371/journal.ppat.1010342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/04/2022] [Accepted: 02/04/2022] [Indexed: 11/18/2022] Open
Abstract
Viral infection of the heart is a common but underappreciated cause of heart failure. Viruses can cause direct cardiac damage by lysing infected cardiomyocytes. Inflammatory immune responses that limit viral replication can also indirectly cause damage during infection, making regulatory factors that fine-tune these responses particularly important. Identifying and understanding these factors that regulate cardiac immune responses during infection will be essential for developing targeted treatments for virus-associated heart failure. Our laboratory has discovered Brain Expressed X-linked protein 1 (BEX1) as a novel stress-regulated pro-inflammatory factor in the heart. Here we report that BEX1 plays a cardioprotective role in the heart during viral infection. Specifically, we adopted genetic gain- and loss-of-function strategies to modulate BEX1 expression in the heart in the context of coxsackievirus B3 (CVB3)-induced cardiomyopathy and found that BEX1 limits viral replication in cardiomyocytes. Interestingly, despite the greater viral load observed in mice lacking BEX1, inflammatory immune cell recruitment in the mouse heart was profoundly impaired in the absence of BEX1. Overall, the absence of BEX1 accelerated CVB3-driven heart failure and pathologic heart remodeling. This result suggests that limiting inflammatory cell recruitment has detrimental consequences for the heart during viral infections. Conversely, transgenic mice overexpressing BEX1 in cardiomyocytes revealed the efficacy of BEX1 for counteracting viral replication in the heart in vivo. We also found that BEX1 retains its antiviral role in isolated cells. Indeed, BEX1 was necessary and sufficient to counteract viral replication in both isolated primary cardiomyocytes and mouse embryonic fibroblasts suggesting a broader applicability of BEX1 as antiviral agent that extended to viruses other than CVB3, including Influenza A and Sendai virus. Mechanistically, BEX1 regulated interferon beta (IFN-β) expression in infected cells. Overall, our study suggests a multifaceted role of BEX1 in the cardiac antiviral immune response.
Collapse
Affiliation(s)
- Colton R. Martens
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Lisa E. Dorn
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Shyam S. Bansal
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
11
|
Akhtar S. Preoperative evaluation of geriatric patients undergoing liver transplantation. Curr Opin Anaesthesiol 2022; 35:96-104. [PMID: 34878418 DOI: 10.1097/aco.0000000000001084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW As the population of the world is aging the number of geriatric patients undergoing liver transplantation (LT) is also increasing. They pose a unique challenge for the caregivers, as they have age-related physiological changes, multiple comorbidities and cirrhosis-related pathologies. RECENT FINDINGS Twenty-two percent of patients who undergo LT are older than 65 years. Many patients suffer from nonalcoholic steatohepatitis (NASH), hepatocellular carcinoma and hepatitis-C virus. Incidence of NASH tends to increase with age, obesity, diabetes and metabolic syndrome. Elderly patients require comprehensive cognitive, cardiac and pulmonary evaluation prior to LT. Cirrhotic cardiomyopathy, hepatopulmonary syndrome, portopulmonary hypertension and frailty are of specific concern. SUMMARY Proportion of elderly patients who are undergoing LT continues to increase. These patients require comprehensive cardiopulmonary and frailty evaluation. Consensus-based practice advisories need to be developed to standardize preoperative evaluation of geriatric patients awaiting LT.
Collapse
Affiliation(s)
- Shamsuddin Akhtar
- Department of Anesthesiology and Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
12
|
Maluleke TT, Millen AME, Michel FS. The effects of estrogen deficiency and aging on myocardial deformation and motion in normotensive female rats. Menopause 2021; 29:89-95. [PMID: 34905750 DOI: 10.1097/gme.0000000000001884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Estrogen deficiency is associated with left ventricular (LV) dysfunction in postmenopausal women and ovariectomized rats. Whether the relationship between estrogen deficiency and LV dysfunction is independent of cardiovascular disease (CVD) risk factors remains uncertain. This study assessed the effects of short-term and long-term estrogen deficiency on cardiac structure and function using conventional and speckle tracking echocardiography, independent of traditional CVD risk factors. METHODS Female Sprague-Dawley rats were divided into short-term (6 wks) ovariectomized (n = 9), short-term sham-operated (n = 10), long-term (6 mo) ovariectomized (n = 8), and long-term sham-operated (n = 9) groups. Cardiac geometry, systolic and diastolic function, and myocardial deformation and motion were measured using echocardiography. RESULTS Ovariectomy had no effect on conventional echocardiography measures of cardiac structure or function. Compared with short-term, long-term groups had reduced LV internal diameter (false discovery rate [FDR] adjusted P = 0.05) and impaired relaxation (e'; FDR adjusted P = 0.0005) independent of body mass and blood pressure (BP). Global longitudinal strain was impaired in ovariectomized compared with sham-operated rats (FDR adjusted P = 0.05), but not after adjusting for body mass and BP (FDR adjusted P = 0.16). Global longitudinal strain (FDR adjusted P = 0.05), strain rate (FDR adjusted P = 0.002), and velocity (FDR adjusted P = 0.04) were impaired in long-term compared with short-term groups. Global longitudinal strain rate remained impaired after adjustments for body mass and BP (FDR adjusted P = 0.02). CONCLUSIONS Estrogen deficiency does not independently cause cardiac remodeling, LV dysfunction, or impaired myocardial deformation. Traditional CVD risk factors accompanying estrogen deficiency may account for cardiac remodeling and dysfunction observed in postmenopausal women.
Collapse
Affiliation(s)
- Tshiamo T Maluleke
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | |
Collapse
|
13
|
Barcena ML, Niehues MH, Christiansen C, Estepa M, Haritonow N, Sadighi AH, Müller-Werdan U, Ladilov Y, Regitz-Zagrosek V. Male Macrophages and Fibroblasts from C57/BL6J Mice Are More Susceptible to Inflammatory Stimuli. Front Immunol 2021; 12:758767. [PMID: 34867999 PMCID: PMC8637417 DOI: 10.3389/fimmu.2021.758767] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Mounting evidence argues for the significant impact of sex in numerous cardiac pathologies, including myocarditis. Macrophage polarization and activation of cardiac fibroblasts play a key role in myocardial inflammation and remodeling. However, the role of sex in these processes is still poorly understood. In this study, we investigated sex-specific alterations in the polarization of murine bone marrow-derived macrophages (BMMs) and the polarization-related changes in fibroblast activation. Cultured male and female murine BMMs from C57/BL6J mice were polarized into M1 (LPS) and M2 (IL-4/IL-13) macrophages. Furthermore, male and female cardiac fibroblasts from C57/BL6J mice were activated with TNF-α, TGF-β, or conditioned medium from M1 BMMs. We found a significant overexpression of M1 markers (c-fos, NFκB, TNF-α, and IL-1β) and M2 markers (MCP-1 and YM1) in male but not female activated macrophages. In addition, the ROS levels were higher in M1 male BMMs, indicating a stronger polarization. Similarly, the pro-fibrotic markers TGF-β and IL-1β were expressed in activated cardiac male fibroblasts at a significantly higher level than in female fibroblasts. In conclusion, the present study provides strong evidence for the male-specific polarization of BMMs and activation of cardiac fibroblasts in an inflammatory environment. The data show an increased inflammatory response and tissue remodeling in male mice.
Collapse
Affiliation(s)
- Maria Luisa Barcena
- Department of Geriatrics and Medical Gerontology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin Partner Site, Berlin, Germany
| | - Maximilian H Niehues
- Department of Geriatrics and Medical Gerontology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Céline Christiansen
- Department of Geriatrics and Medical Gerontology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Misael Estepa
- Department of Geriatrics and Medical Gerontology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Internal Medicine and Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Natalie Haritonow
- Department of Geriatrics and Medical Gerontology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Amir H Sadighi
- Department of Geriatrics and Medical Gerontology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ursula Müller-Werdan
- Department of Geriatrics and Medical Gerontology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Yury Ladilov
- Institute for Gender in Medicine, Center for Cardiovascular Research, Charité University Hospital, Berlin, Germany
| | - Vera Regitz-Zagrosek
- Institute for Gender in Medicine, Center for Cardiovascular Research, Charité University Hospital, Berlin, Germany.,Department of Cardiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| |
Collapse
|
14
|
Khan A, Ramos-Gomes F, Markus A, Mietsch M, Hinkel R, Alves F. Label-free imaging of age-related cardiac structural changes in non-human primates using multiphoton nonlinear microscopy. BIOMEDICAL OPTICS EXPRESS 2021; 12:7009-7023. [PMID: 34858695 PMCID: PMC8606147 DOI: 10.1364/boe.432102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 06/13/2023]
Abstract
Heart failure is one of the most common causes of morbidity and mortality. Both maturational abnormalities and age-associated cardiac pathologies contribute to heart failure. Imaging-based assessment to discern detailed cardiac structure at various maturational stages is imperative for understanding mechanisms behind cardiac growth and aging. Using multiphoton nonlinear optical microscopy (NLOM) based label-free imaging, we investigated cardiac structural composition in a human-relevant aging model, the common marmoset monkey (Callithrix jacchus). Animals were divided into three different age groups including neonatal, young adult and old. By devising a unique strategy for segregating collagen and myosin emitted second harmonic generation (SHG) signals, we performed a volumetric assessment of collagen and total scattering tissue (collagen + myosin). Aged marmoset hearts exhibited an increase in collagen and total scattering tissue volume at the sites of severe tissue remodelling indicating age-related cardiac fibrosis. Significantly low scattering tissue volume in neonatal marmoset hearts was attributed to a lack of binding between the myofibrils in maturing cardiac tissue. Comprehensive quantitative assessment of structural composition during maturation and aging of marmoset hearts revealed significant differences in myofibril length, alignment, curvature and angular distribution. In conclusion, label-free high-resolution NLOM facilitates visualization and quantification of subcellular structural features for understanding vital age-related morphological alterations in the marmoset heart.
Collapse
Affiliation(s)
- Amara Khan
- Max-Planck-Institute for Experimental Medicine, Translational Molecular Imaging, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, 37077 Göttingen, Germany
| | - Fernanda Ramos-Gomes
- Max-Planck-Institute for Experimental Medicine, Translational Molecular Imaging, Göttingen, Germany
| | - Andrea Markus
- Max-Planck-Institute for Experimental Medicine, Translational Molecular Imaging, Göttingen, Germany
| | - Matthias Mietsch
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, 37077 Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Rabea Hinkel
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, 37077 Göttingen, Germany
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- Stiftung Tierärztliche Hochschule Hannover, Hannover, Germany
| | - Frauke Alves
- Max-Planck-Institute for Experimental Medicine, Translational Molecular Imaging, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, 37077 Göttingen, Germany
- University Medical Center Göttingen, Institute for Diagnostic and Interventional Radiology & Clinic for Hematology and Medical Oncology, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells,” Göttingen, Germany
| |
Collapse
|
15
|
Moura B, Aimo A, Al-Mohammad A, Flammer A, Barberis V, Bayes-Genis A, Brunner-La Rocca HP, Fontes-Carvalho R, Grapsa J, Hülsmann M, Ibrahim N, Knackstedt C, Januzzi JL, Lapinskas T, Sarrias A, Matskeplishvili S, Meijers WC, Messroghli D, Mueller C, Pavo N, Simonavičius J, Teske AJ, van Kimmenade R, Seferovic P, Coats AJS, Emdin M, Richards AM. Integration of imaging and circulating biomarkers in heart failure: a consensus document by the Biomarkers and Imaging Study Groups of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2021; 23:1577-1596. [PMID: 34482622 DOI: 10.1002/ejhf.2339] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/28/2021] [Accepted: 08/29/2021] [Indexed: 12/28/2022] Open
Abstract
Circulating biomarkers and imaging techniques provide independent and complementary information to guide management of heart failure (HF). This consensus document by the Heart Failure Association (HFA) of the European Society of Cardiology (ESC) presents current evidence-based indications relevant to integration of imaging techniques and biomarkers in HF. The document first focuses on application of circulating biomarkers together with imaging findings, in the broad domains of screening, diagnosis, risk stratification, guidance of treatment and monitoring, and then discusses specific challenging settings. In each section we crystallize clinically relevant recommendations and identify directions for future research. The target readership of this document includes cardiologists, internal medicine specialists and other clinicians dealing with HF patients.
Collapse
Affiliation(s)
- Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiology Department, Porto Armed Forces Hospital, Porto, Portugal
| | - Alberto Aimo
- Scuola Superiore Sant'Anna, and Fondazione G. Monasterio, Pisa, Italy
| | - Abdallah Al-Mohammad
- Medical School, University of Sheffield and Sheffield Teaching Hospitals, Sheffield, UK
| | | | | | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Hans-Peter Brunner-La Rocca
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ricardo Fontes-Carvalho
- Cardiovascular Research and Development Unit (UnIC), Faculty of Medicine University of Porto, Porto, Portugal.,Cardiology Department, Centro Hospitalar de Vila Nova Gaia/Espinho, Espinho, Portugal
| | - Julia Grapsa
- Department of Cardiology, Guys and St Thomas NHS Hospitals Trust, London, UK
| | - Martin Hülsmann
- Department of Internal Medicine, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Nasrien Ibrahim
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christian Knackstedt
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomas Lapinskas
- Department of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Axel Sarrias
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | | | - Daniel Messroghli
- Department of Internal Medicine-Cardiology, Deutsches Herzzentrum Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Mueller
- Department of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Noemi Pavo
- Department of Internal Medicine, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Justas Simonavičius
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.,Vilnius University Hospital Santaros klinikos, Vilnius, Lithuania
| | - Arco J Teske
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roland van Kimmenade
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | | | - Michele Emdin
- Scuola Superiore Sant'Anna, and Fondazione G. Monasterio, Pisa, Italy
| | - A Mark Richards
- Christchurch Heart Institute, University of Otago, Dunedin, New Zealand.,Cardiovascular Research Institute, National University of Singapore, Singapore
| |
Collapse
|
16
|
Bou-Teen D, Kaludercic N, Weissman D, Turan B, Maack C, Di Lisa F, Ruiz-Meana M. Mitochondrial ROS and mitochondria-targeted antioxidants in the aged heart. Free Radic Biol Med 2021; 167:109-124. [PMID: 33716106 DOI: 10.1016/j.freeradbiomed.2021.02.043] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/14/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
Excessive mitochondrial ROS production has been causally linked to the pathophysiology of aging in the heart and other organs, and plays a deleterious role in several age-related cardiac pathologies, including myocardial ischemia-reperfusion injury and heart failure, the two worldwide leading causes of death and disability in the elderly. However, ROS generation is also a fundamental mitochondrial function that orchestrates several signaling pathways, some of them exerting cardioprotective effects. In cardiac myocytes, mitochondria are particularly abundant and are specialized in subcellular populations, in part determined by their relationships with other organelles and their cyclic calcium handling activity necessary for adequate myocardial contraction/relaxation and redox balance. Depending on their subcellular location, mitochondria can themselves be differentially targeted by ROS and display distinct age-dependent functional decline. Thus, precise mitochondria-targeted therapies aimed at counteracting unregulated ROS production are expected to have therapeutic benefits in certain aging-related heart conditions. However, for an adequate design of such therapies, it is necessary to unravel the complex and dynamic interactions between mitochondria and other cellular processes.
Collapse
Affiliation(s)
- Diana Bou-Teen
- Hospital Universitari Vall d'Hebron, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR),Universitat Autonoma de Barcelona, 08035, Barcelona, Spain
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), via Ugo Bassi 58/B, 35131, Padova, Italy; Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35129, Padova, Italy
| | - David Weissman
- Comprehensive Heart Failure Center, University Clinic Würzburg, 97080, Würzburg, Germany
| | - Belma Turan
- Departments of Biophysics, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, 97080, Würzburg, Germany
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR), via Ugo Bassi 58/B, 35131, Padova, Italy; Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Marisol Ruiz-Meana
- Hospital Universitari Vall d'Hebron, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR),Universitat Autonoma de Barcelona, 08035, Barcelona, Spain; Centro de Investigación Biomédica en Red-CV, CIBER-CV, Spain.
| |
Collapse
|
17
|
Abstract
ABSTRACT As an integral component of cardiac tissue, macrophages are critical for cardiac development, adult heart homeostasis, as well as cardiac healing. One fundamental function of macrophages involves the clearance of dying cells or debris, a process termed efferocytosis. Current literature primarily pays attention to the impact of efferocytosis on apoptotic cells. However, emerging evidence suggests that necrotic cells and their released cellular debris can also be removed by cardiac macrophages through efferocytosis. Importantly, recent studies have demonstrated that macrophage efferocytosis plays an essential role in cardiac pathophysiology and repair. Therefore, understanding macrophage efferocytosis would provide valuable insights on cardiac health, and may offer new therapeutic strategies for the treatment of patients with heart failure. In this review, we first summarize the molecular signals that are associated with macrophage efferocytosis of apoptotic and necrotic cells, and then discuss how the linkage of efferocytosis to the resolution of inflammation affects cardiac function and recovery under normal and diseased conditions. Lastly, we highlight new discoveries related to the effects of macrophage efferocytosis on cardiac injury and repair.
Collapse
Affiliation(s)
- Li Yutian
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Li Qianqian
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Pharmaceutical Science, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
18
|
Sparks MA, Rianto F, Diaz E, Revoori R, Hoang T, Bouknight L, Stegbauer J, Vivekanandan-Giri A, Ruiz P, Pennathur S, Abraham DM, Gurley SB, Crowley SD, Coffman TM. Direct Actions of AT 1 (Type 1 Angiotensin) Receptors in Cardiomyocytes Do Not Contribute to Cardiac Hypertrophy. Hypertension 2021; 77:393-404. [PMID: 33390039 PMCID: PMC7803456 DOI: 10.1161/hypertensionaha.119.14079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text. Activation of AT1 (type 1 Ang) receptors stimulates cardiomyocyte hypertrophy in vitro. Accordingly, it has been suggested that regression of cardiac hypertrophy associated with renin-Ang system blockade is due to inhibition of cellular actions of Ang II in the heart, above and beyond their effects to reduce pressure overload. We generated 2 distinct mouse lines with cell-specific deletion of AT1A receptors, from cardiomyocytes. In the first line (C-SMKO), elimination of AT1A receptors was achieved using a heterologous Cre recombinase transgene under control of the Sm22 promoter, which expresses in cells of smooth muscle lineage including cardiomyocytes and vascular smooth muscle cells of conduit but not resistance vessels. The second line (R-SMKO) utilized a Cre transgene knocked-in to the Sm22 locus, which drives expression in cardiac myocytes and vascular smooth muscle cells in both conduit and resistance arteries. Thus, although both groups lack AT1 receptors in the cardiomyocytes, they are distinguished by presence (C-SMKO) or absence (R-SMKO) of peripheral vascular responses to Ang II. Similar to wild-types, chronic Ang II infusion caused hypertension and cardiac hypertrophy in C-SMKO mice, whereas both hypertension and cardiac hypertrophy were reduced in R-SMKOs. Thus, despite the absence of AT1A receptors in cardiomyocytes, C-SMKOs develop robust cardiac hypertrophy. By contrast, R-SMKOs developed identical levels of hypertrophy in response to pressure overload–induced by transverse aortic banding. Our findings suggest that direct activation of AT1 receptors in cardiac myocytes has minimal influence on cardiac hypertrophy induced by renin-Ang system activation or pressure overload.
Collapse
Affiliation(s)
- Matthew A Sparks
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC.,Renal Section, Durham VA Health System, NC (M.A.S, S.D.C., T.M.C.)
| | - Fitra Rianto
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC
| | - Edward Diaz
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC
| | - Ritika Revoori
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC
| | - Thien Hoang
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC
| | - Lucas Bouknight
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC
| | - Johannes Stegbauer
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC.,Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Germany (J.S.)
| | - Anuradha Vivekanandan-Giri
- Division of Nephrology, Department of Medicine, Michigan University Medical Center, Ann Arbor (A.V.-G., S.P.)
| | - Phillip Ruiz
- Department of Surgery and Pathology, University of Miami, FL (P.R.)
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Medicine, Michigan University Medical Center, Ann Arbor (A.V.-G., S.P.)
| | - Dennis M Abraham
- Division of Cardiology, Department of Medicine (D.M.A.), Duke University School of Medicine, Durham, NC
| | - Susan B Gurley
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Sciences University, Portland (S.B.G.)
| | - Steven D Crowley
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC.,Renal Section, Durham VA Health System, NC (M.A.S, S.D.C., T.M.C.)
| | - Thomas M Coffman
- From the Division of Nephrology, Department of Medicine (M.A.S., F.R., E.D., R.R., T.H., L.B., J.S., S.D.C., T.M.C.), Duke University School of Medicine, Durham, NC.,Renal Section, Durham VA Health System, NC (M.A.S, S.D.C., T.M.C.).,Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore (T.M.C.)
| |
Collapse
|
19
|
Díez J, González A, Kovacic JC. Myocardial Interstitial Fibrosis in Nonischemic Heart Disease, Part 3/4: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:2204-2218. [PMID: 32354386 DOI: 10.1016/j.jacc.2020.03.019] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/17/2022]
Abstract
Myocardial interstitial fibrosis (MIF) is a histological hallmark of several cardiac diseases that alter myocardial architecture and function and are associated with progression to heart failure. MIF is a diffuse and patchy process, appearing as a combination of interstitial microscars, perivascular collagen fiber deposition, and increased thickness of mysial collagen strands. Although MIF arises mainly because of alterations in fibrillar collagen turnover leading to collagen fiber accumulation, there are also alterations in other nonfibrillar extracellular matrix components, such as fibronectin and matricellular proteins. Furthermore, in addition to an excess of collagen, qualitative changes in collagen fibers also contribute to the detrimental impact of MIF. In this part 3 of a 4-part JACC Focus Seminar, we review the evidence on the complex mechanisms leading to MIF, as well as its contribution to systolic and diastolic cardiac dysfunction and impaired clinical outcomes in patients with nonischemic heart disease.
Collapse
Affiliation(s)
- Javier Díez
- Program of Cardiovascular Diseases, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain; Department of Nephrology, University of Navarra Clinic, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red-Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, Madrid, Spain.
| | - Arantxa González
- Program of Cardiovascular Diseases, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red-Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, Madrid, Spain
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
20
|
JavaCyte, a novel open-source tool for automated quantification of key hallmarks of cardiac structural remodeling. Sci Rep 2020; 10:20074. [PMID: 33208780 PMCID: PMC7675975 DOI: 10.1038/s41598-020-76932-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/24/2020] [Indexed: 11/08/2022] Open
Abstract
Many cardiac pathologies involve changes in tissue structure. Conventional analysis of structural features is extremely time-consuming and subject to observer bias. The possibility to determine spatial interrelations between these features is often not fully exploited. We developed a staining protocol and an ImageJ-based tool (JavaCyte) for automated histological analysis of cardiac structure, including quantification of cardiomyocyte size, overall and endomysial fibrosis, spatial patterns of endomysial fibrosis, fibroblast density, capillary density and capillary size. This automated analysis was compared to manual quantification in several well-characterized goat models of atrial fibrillation (AF). In addition, we tested inter-observer variability in atrial biopsies from the CATCH-ME consortium atrial tissue bank, with patients stratified by their cardiovascular risk profile for structural remodeling. We were able to reproduce previous manually derived histological findings in goat models for AF and AV block (AVB) using JavaCyte. Furthermore, strong correlation was found between manual and automated observations for myocyte count (r = 0.94, p < 0.001), myocyte diameter (r = 0.97, p < 0.001), endomysial fibrosis (r = 0.98, p < 0.001) and capillary count (r = 0.95, p < 0.001) in human biopsies. No significant variation between observers was observed (ICC = 0.89, p < 0.001). We developed and validated an open-source tool for high-throughput, automated histological analysis of cardiac tissue properties. JavaCyte was as accurate as manual measurements, with less inter-observer variability and faster throughput.
Collapse
|
21
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
22
|
Recinella L, Orlando G, Ferrante C, Chiavaroli A, Brunetti L, Leone S. Adipokines: New Potential Therapeutic Target for Obesity and Metabolic, Rheumatic, and Cardiovascular Diseases. Front Physiol 2020; 11:578966. [PMID: 33192583 PMCID: PMC7662468 DOI: 10.3389/fphys.2020.578966] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Besides its role as an energy storage organ, adipose tissue can be viewed as a dynamic and complex endocrine organ, which produces and secretes several adipokines, including hormones, cytokines, extracellular matrix (ECM) proteins, and growth and vasoactive factors. A wide body of evidence showed that adipokines play a critical role in various biological and physiological functions, among which feeding modulation, inflammatory and immune function, glucose and lipid metabolism, and blood pressure control. The aim of this review is to summarize the effects of several adipokines, including leptin, diponectin, resistin, chemerin, lipocalin-2 (LCN2), vaspin, omentin, follistatin-like 1 (FSTL1), secreted protein acidic and rich in cysteine (SPARC), secreted frizzled-related protein 5 (SFRP5), C1q/TNF-related proteins (CTRPs), family with sequence similarity to 19 member A5 (FAM19A5), wingless-type inducible signaling pathway protein-1 (WISP1), progranulin (PGRN), nesfatin-1 (nesfatin), visfatin/PBEF/NAMPT, apelin, retinol binding protein 4 (RPB4), and plasminogen activator inhibitor-1 (PAI-1) in the regulation of insulin resistance and vascular function, as well as many aspects of inflammation and immunity and their potential role in managing obesity-associated diseases, including metabolic, osteoarticular, and cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | - Luigi Brunetti
- Department of Pharmacy, Gabriele d’Annunzio University, Chieti, Italy
| | | |
Collapse
|
23
|
Effects of Quercetin on Cardiac Function in Pressure Overload and Postischemic Cardiac Injury in Rodents: a Systematic Review and Meta-Analysis. Cardiovasc Drugs Ther 2020; 36:15-29. [PMID: 33064235 DOI: 10.1007/s10557-020-07100-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2020] [Indexed: 12/28/2022]
Abstract
PURPOSE Cardiac dysfunction can occur as a sequela of a state of prolonged pressure overload and postischemic injury. Flavonoids such as quercetin may be protective against cardiovascular disease. This study aimed to systematically assess the effects of quercetin on cardiac function in pressure overload and postischemia-reperfusion injury in rodents. METHODS A systematic search of the literature up to May 2020 was conducted in PubMed, Ovid Medline, EBSCOhost, Scopus, and the Cochrane Library to identify relevant published studies on quercetin and cardiac function using standardized criteria. Meta-analyses were performed on animal studies of pressure overload and ischemia-reperfusion (I/R) injury. RESULTS The effects of quercetin on cardiac function in both models were qualitatively reported in 14 studies. The effects of quercetin in four pressure-overload model studies involving 73 rodents and eight I/R-injury model studies involving 120 rodents were quantitatively assessed by meta-analysis. Quercetin improved the overall cardiac function in both pressure overload (n = 4 studies, n = 73 rodents; SMD = - 1.50; 95% CI: - 2.66 to - 0.33; P < 0.05; I2 = 74.05%) and I/R injury (n = 8 studies, n = 120 rodents; SMD = - 1.81; 95% CI: - 3.05 to - 0.56; P < 0.01; I2 = 84.93%) models. The improvement was associated with amelioration in cardiac structure in the pressure-overload model and both systolic and diastolic functioning in the I/R-injury model. CONCLUSION The present meta-analysis suggested that quercetin has beneficial effects for improving cardiac left ventricular dysfunction in both pressure-overload and I/R-injury models.
Collapse
|
24
|
Keshavarz-Bahaghighat H, Darwesh AM, Sosnowski DK, Seubert JM. Mitochondrial Dysfunction and Inflammaging in Heart Failure: Novel Roles of CYP-Derived Epoxylipids. Cells 2020; 9:E1565. [PMID: 32604981 PMCID: PMC7408578 DOI: 10.3390/cells9071565] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 02/07/2023] Open
Abstract
Age-associated changes leading to a decline in cardiac structure and function contribute to the increased susceptibility and incidence of cardiovascular diseases (CVD) in elderly individuals. Indeed, age is considered a risk factor for heart failure and serves as an important predictor for poor prognosis in elderly individuals. Effects stemming from chronic, low-grade inflammation, inflammaging, are considered important determinants in cardiac health; however, our understanding of the mechanisms involved remains unresolved. A steady decline in mitochondrial function is recognized as an important biological consequence found in the aging heart which contributes to the development of heart failure. Dysfunctional mitochondria contribute to increased cellular stress and an innate immune response by activating the NLRP-3 inflammasomes, which have a role in inflammaging and age-related CVD pathogenesis. Emerging evidence suggests a protective role for CYP450 epoxygenase metabolites of N-3 and N-6 polyunsaturated fatty acids (PUFA), epoxylipids, which modulate various aspects of the immune system and protect mitochondria. In this article, we provide insight into the potential roles N-3 and N-6 PUFA have modulating mitochondria, inflammaging and heart failure.
Collapse
Affiliation(s)
- Hedieh Keshavarz-Bahaghighat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
| | - Ahmed M. Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
| | - Deanna K. Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta 2020-M Katz Group Centre for Pharmacy and Health Research 11361-87 Avenue, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
25
|
O'Brien M, Baicu CF, Van Laer AO, Zhang Y, McDonald LT, LaRue AC, Zile MR, Bradshaw AD. Pressure overload generates a cardiac-specific profile of inflammatory mediators. Am J Physiol Heart Circ Physiol 2020; 319:H331-H340. [PMID: 32589444 DOI: 10.1152/ajpheart.00274.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Mechanisms that contribute to myocardial fibrosis, particularly in response to left ventricular pressure overload (LVPO), remain poorly defined. To test the hypothesis that a myocardial-specific profile of secreted factors is produced in response to PO, levels of 44 factors implicated in immune cell recruitment and function were assessed in a murine model of cardiac hypertrophy and compared with levels produced in a model of pulmonary fibrosis (PF). Mice subjected to PO were assessed at 1 and 4 wk. Protein from plasma, LV, lungs, and kidneys were analyzed by specific protein array analysis in parallel with protein from mice subjected to silica-instilled PF. Of the 44 factors assessed, 13 proteins were elevated in 1-wk PO myocardium, whereas 18 proteins were found increased in fibrotic lung. Eight of those increased in 1-wk LVPO were not found to be increased in fibrotic lungs (CCL-11, CCL-12, CCL-17, CCL-19, CCL-21, CCL-22, IL-16, and VEGF). Additionally, six factors were increased in plasma of 1-wk LVPO in the absence of increases in myocardial levels. In contrast, in mice with PF, no factors were found increased in plasma that were not elevated in lung tissue. Of those factors increased at 1 wk, only TIMP-1 remained elevated at 4 wk of LVPO. Immunohistochemistry of myocardial vasculature at 1 and 4 wk revealed similar amounts of total vasculature; however, evidence of activated endothelium was observed at 1 wk and, to a lesser extent, at 4 wk LVPO. In conclusion, PO myocardium generated a unique signature of cytokine expression versus that of fibrotic lung.NEW & NOTEWORTHY Myocardial fibrosis and the resultant increases in myocardial stiffness represent pivotal consequences of chronic pressure overload (PO). In this study, cytokine profiles produced in a murine model of cardiac fibrosis induced by PO were compared with those produced in response to silica-induced lung fibrosis. A unique profile of cardiac tissue-specific and plasma-derived factors generated in response to PO are reported.
Collapse
Affiliation(s)
- Matthew O'Brien
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Catalin F Baicu
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - An O Van Laer
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Yuhua Zhang
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Lindsey T McDonald
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Amanda C LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Michael R Zile
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Amy D Bradshaw
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
26
|
Blackwood EA, Bilal AS, Azizi K, Sarakki A, Glembotski CC. Simultaneous Isolation and Culture of Atrial Myocytes, Ventricular Myocytes, and Non-Myocytes from an Adult Mouse Heart. J Vis Exp 2020:10.3791/61224. [PMID: 32597844 PMCID: PMC8580476 DOI: 10.3791/61224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The isolation and culturing of cardiac myocytes from mice has been essential for furthering the understanding of cardiac physiology and pathophysiology. While isolating myocytes from neonatal mouse hearts is relatively straightforward, myocytes from the adult murine heart are preferred. This is because compared to neonatal cells, adult myocytes more accurately recapitulate cell function as it occurs in the adult heart in vivo. However, it is technically difficult to isolate adult mouse cardiac myocytes in the necessary quantities and viability, which contributes to an experimental impasse. Furthermore, published procedures are specific for the isolation of either atrial or ventricular myocytes at the expense of atrial and ventricular non-myocyte cells. Described here is a detailed method for isolating both atrial and ventricular cardiac myocytes, along with atrial and ventricular non-myocytes, simultaneously from a single mouse heart. Also provided are the details for optimal cell-specific culturing methods, which enhance cell viability and function. This protocol aims not only to expedite the process of adult murine cardiac cell isolation, but also to increase the yield and viability of cells for investigations of atrial and ventricular cardiac cells.
Collapse
Affiliation(s)
- Erik A Blackwood
- San Diego State University Heart Institute and the Department of Biology, San Diego State University
| | - Alina S Bilal
- San Diego State University Heart Institute and the Department of Biology, San Diego State University
| | - Khalid Azizi
- San Diego State University Heart Institute and the Department of Biology, San Diego State University
| | - Anup Sarakki
- San Diego State University Heart Institute and the Department of Biology, San Diego State University
| | - Christopher C Glembotski
- San Diego State University Heart Institute and the Department of Biology, San Diego State University;
| |
Collapse
|
27
|
Sharifi BG, Yang M, Shah PK. Aging and GATA3-positive macrophages. Aging (Albany NY) 2020; 11:2179-2180. [PMID: 31017875 PMCID: PMC6520000 DOI: 10.18632/aging.101929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 04/19/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Behrooz G Sharifi
- Oppenheimer Atherosclerosis Research Center, Cedars Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mingjie Yang
- Oppenheimer Atherosclerosis Research Center, Cedars Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Prediman K Shah
- Oppenheimer Atherosclerosis Research Center, Cedars Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
28
|
Tracy E, Rowe G, LeBlanc AJ. Cardiac tissue remodeling in healthy aging: the road to pathology. Am J Physiol Cell Physiol 2020; 319:C166-C182. [PMID: 32432929 DOI: 10.1152/ajpcell.00021.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review aims to highlight the normal physiological remodeling that occurs in healthy aging hearts, including changes that occur in contractility, conduction, valve function, large and small coronary vessels, and the extracellular matrix. These "normal" age-related changes serve as the foundation that supports decreased plasticity and limited ability for tissue remodeling during pathophysiological states such as myocardial ischemia and heart failure. This review will identify populations at greater risk for poor tissue remodeling in advanced age along with present and future therapeutic strategies that may ameliorate dysfunctional tissue remodeling in aging hearts.
Collapse
Affiliation(s)
- Evan Tracy
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | - Amanda J LeBlanc
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| |
Collapse
|
29
|
Unveiling the Role of Inflammation and Oxidative Stress on Age-Related Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1954398. [PMID: 32454933 PMCID: PMC7232723 DOI: 10.1155/2020/1954398] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/12/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022]
Abstract
The global population above 60 years has been growing exponentially in the last decades, which is accompanied by an increase in the prevalence of age-related chronic diseases, highlighting cardiovascular diseases (CVDs), such as hypertension, atherosclerosis, and heart failure. Aging is the main risk factor for these diseases. Such susceptibility to disease is explained, at least in part, by the increase of oxidative stress, in which it damages cellular components such as proteins, DNA, and lipids. In addition, the chronic inflammatory process in aging “inflammaging” also contributes to cell damage, creating a stressful environment which drives to the development of CVDs. Taken together, it is possible to identify the molecular connection between oxidative stress and the inflammatory process, especially by the crosstalk between the transcription factors Nrf-2 and NF-κB which are mediated by redox signalling and are involved in aging. Therapies that control this process are key targets in the prevention/combat of age-related CVDs. In this review, we show the basics of inflammation and oxidative stress, including the crosstalk between them, and the implications on age-related CVDs.
Collapse
|
30
|
Neutrophil-Lymphocyte Ratio in Patients with Acute Heart Failure Predicts In-Hospital and Long-Term Mortality. J Clin Med 2020; 9:jcm9020557. [PMID: 32085596 PMCID: PMC7073552 DOI: 10.3390/jcm9020557] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 01/18/2023] Open
Abstract
The application of a simple blood test to predict prognosis in acute heart failure (AHF) patients is not well established. Neutrophil-lymphocyte ratio (NLR) is inexpensive and easy to obtain in hospitalized patients using a routine blood test. We evaluate the prognostic implications of NLR as an independent predictor of in-hospital and long-term mortality in AHF patients. Among 5625 patients enrolled in the Korean Acute Heart Failure registry, 5580 patients were classified into quartiles by their NLR level, and analyzed for in-hospital and post-discharge three-year mortality. Patients in the highest NLR quartile had the highest in-hospital and post-discharge three-year mortality. The same results were seen by dividing the aggravating factor into the infection or ischemia group and the non-infection or non-ischemia group. For patients aggravated from infection or ischemia, a cut-off NLR value was 7.0 that increase the risk of in-hospital and post-discharge three-year mortality. In subgroups of patients not aggravated from infection or ischemia, a cut-off NLR value was 5.0 that increase the risk of in-hospital and post discharge three-year mortality. Elevated NLR in AHF patients at the index hospitalization is an independent predictor for in-hospital and post-discharge three-year mortality. Taken together, NLR is a marker for risk assessment of AHF patients.
Collapse
|
31
|
Serna E, Mastaloudis A, Martorell P, Wood SM, Hester SN, Bartlett M, Prolla TA, Viña J. A Novel Micronutrient Blend Mimics Calorie Restriction Transcriptomics in Multiple Tissues of Mice and Increases Lifespan and Mobility in C. elegans. Nutrients 2020; 12:nu12020486. [PMID: 32075050 PMCID: PMC7071149 DOI: 10.3390/nu12020486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 11/17/2022] Open
Abstract
Background: We previously described a novel micronutrient blend that behaves like a putative calorie restriction mimetic. The aim of this paper was to analyze the beneficial effects of our micronutrient blend in mice and C. elegans, and compare them with calorie restriction. Methods: Whole transcriptomic analysis was performed in the brain cortex, skeletal muscle and heart in three groups of mice: old controls (30 months), old + calorie restriction and old + novel micronutrient blend. Longevity and vitality were tested in C. elegans. Results: The micronutrient blend elicited transcriptomic changes in a manner similar to those in the calorie-restricted group and different from those in the control group. Subgroup analysis revealed that nuclear hormone receptor, proteasome complex and angiotensinogen genes, all of which are known to be directly related to aging, were the most affected. Furthermore, a functional analysis in C. elegans was used. We found that feeding C. elegans the micronutrient blend increased longevity as well as vitality. Conclusions: We describe a micronutrient supplement that causes similar changes (transcriptomic and promoting longevity and vitality) as a calorie restriction in mice and C. elegans, respectively, but further studies are required to confirm these effects in humans.
Collapse
Affiliation(s)
- Eva Serna
- Freshage Research Group-Dept. Physiology-University of Valencia, CIBERFES, INCLIVA, 46010 Valencia, Spain;
| | - Angela Mastaloudis
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Patricia Martorell
- Cell Biology Laboratory/ADM Nutrition/Biopolis SL/Archer Daniels Midland, 46980 Paterna, Valencia, Spain;
| | - Steven M. Wood
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Shelly N. Hester
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Mark Bartlett
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Tomas A. Prolla
- LifeGen Technologies LLC, Madison, WI 53719, USA;
- Departments of Genetics and Medical Genetics; University of Wisconsin; Madison, WI 53706, USA
| | - Jose Viña
- Freshage Research Group-Dept. Physiology-University of Valencia, CIBERFES, INCLIVA, 46010 Valencia, Spain;
- Correspondence: ; Tel.: +34-963864650
| |
Collapse
|
32
|
Vidal R, Wagner JUG, Braeuning C, Fischer C, Patrick R, Tombor L, Muhly-Reinholz M, John D, Kliem M, Conrad T, Guimarães-Camboa N, Harvey R, Dimmeler S, Sauer S. Transcriptional heterogeneity of fibroblasts is a hallmark of the aging heart. JCI Insight 2019; 4:131092. [PMID: 31723062 DOI: 10.1172/jci.insight.131092] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/02/2019] [Indexed: 12/27/2022] Open
Abstract
Aging is a major risk factor for cardiovascular disease. Although the impact of aging has been extensively studied, little is known regarding the aging processes in cells of the heart. Here we analyzed the transcriptomes of hearts of 12-week-old and 18-month-old mice by single-nucleus RNA-sequencing. Among all cell types, aged fibroblasts showed most significant differential gene expression, increased RNA dynamics, and network entropy. Aged fibroblasts exhibited significantly changed expression patterns of inflammatory, extracellular matrix organization angiogenesis, and osteogenic genes. Functional analyses indicated deterioration of paracrine signatures between fibroblasts and endothelial cells in old hearts. Aged heart-derived fibroblasts had impaired endothelial cell angiogenesis and autophagy and augmented proinflammatory response. In particular, expression of Serpine1 and Serpine2 were significantly increased and secreted by old fibroblasts to exert antiangiogenic effects on endothelial cells, an effect that could be significantly prevented by using neutralizing antibodies. Moreover, we found an enlarged subpopulation of aged fibroblasts expressing osteoblast genes in the epicardial layer associated with increased calcification. Taken together this study provides system-wide insights and identifies molecular changes of aging cardiac fibroblasts, which may contribute to declined heart function.
Collapse
Affiliation(s)
- Ramon Vidal
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Julian Uwe Gabriel Wagner
- Faculty for Biological Sciences and.,Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | | | | | - Ralph Patrick
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales Sydney, Kensington, New South Wales, Australia
| | - Lukas Tombor
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Marion Muhly-Reinholz
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - David John
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Magdalena Kliem
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Thomas Conrad
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Nuno Guimarães-Camboa
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Richard Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales Sydney, Kensington, New South Wales, Australia.,School of Biotechnology and Biomolecular Science, University of New South Wales Sydney, Kensington, New South Wales, Australia
| | - Stefanie Dimmeler
- Faculty for Biological Sciences and.,Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,German Centre for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
33
|
Sawaki D, Czibik G, Pini M, Ternacle J, Suffee N, Mercedes R, Marcelin G, Surenaud M, Marcos E, Gual P, Clément K, Hue S, Adnot S, Hatem SN, Tsuchimochi I, Yoshimitsu T, Hénégar C, Derumeaux G. Visceral Adipose Tissue Drives Cardiac Aging Through Modulation of Fibroblast Senescence by Osteopontin Production. Circulation 2019; 138:809-822. [PMID: 29500246 DOI: 10.1161/circulationaha.117.031358] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aging induces cardiac structural and functional changes linked to the increased deposition of extracellular matrix proteins, including OPN (osteopontin), conducing to progressive interstitial fibrosis. Although OPN is involved in various pathological conditions, its role in myocardial aging remains unknown. METHODS OPN deficient mice (OPN-/-) with their wild-type (WT) littermates were evaluated at 2 and 14 months of age in terms of cardiac structure, function, histology and key molecular markers. OPN expression was determined by reverse-transcription polymerase chain reaction, immunoblot and immunofluorescence. Luminex assays were performed to screen plasma samples for various cytokines/adipokines in addition to OPN. Similar explorations were conducted in aged WT mice after surgical removal of visceral adipose tissue (VAT) or treatment with a small-molecule OPN inhibitor agelastatin A. Primary WT fibroblasts were incubated with plasma from aged WT and OPN-/- mice, and evaluated for senescence (senescence-associated β-galactosidase and p16), as well as fibroblast activation markers (Acta2 and Fn1). RESULTS Plasma OPN levels increased in WT mice during aging, with VAT showing the strongest OPN induction contrasting with myocardium that did not express OPN. VAT removal in aged WT mice restored cardiac function and decreased myocardial fibrosis in addition to a substantial reduction of circulating OPN and transforming growth factor β levels. OPN deficiency provided a comparable protection against age-related cardiac fibrosis and dysfunction. Intriguingly, a strong induction of senescence in cardiac fibroblasts was observed in both VAT removal and OPN-/- mice. The addition of plasma from aged OPN-/- mice to cultures of primary cardiac fibroblasts induced senescence and reduced their activation (compared to aged WT plasma). Finally, Agelastatin A treatment of aged WT mice fully reversed age-related myocardial fibrosis and dysfunction. CONCLUSIONS During aging, VAT represents the main source of OPN and alters heart structure and function via its profibrotic secretome. As a proof-of-concept, interventions targeting OPN, such as VAT removal and OPN deficiency, rescued the heart and induced a selective modulation of fibroblast senescence. Our work uncovers OPN's role in the context of myocardial aging and proposes OPN as a potential new therapeutic target for a healthy cardiac aging.
Collapse
Affiliation(s)
- Daigo Sawaki
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Gabor Czibik
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Maria Pini
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Julien Ternacle
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP, Department of Cardiology, Henri Mondor Hospital, DHU-ATVB (J.T., G.D.)
| | - Nadine Suffee
- Sorbonne Université, INSERM UMRS 1166, Institute of Cardiometabolism and Nutrition ICAN (N.S., S.H.)
| | - Raquel Mercedes
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Geneviève Marcelin
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital (G.M., K.C.)
- Sorbonne Universities, Université Pierre et Marie Curie, University of Paris 06, INSERM UMR_S 1166, Nutriomics Team 6 (G.M., K.C.)
| | - Mathieu Surenaud
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP Vaccine Research Institute (VRI) (M.S., S.H.)
| | - Elisabeth Marcos
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Philippe Gual
- INSERM, U1065, C3M, Team 8 "hepatic complications in obesity" (P.G.)
- Université Côte d'Azur (P.G.)
| | - Karine Clément
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital (G.M., K.C.)
- Sorbonne Universities, Université Pierre et Marie Curie, University of Paris 06, INSERM UMR_S 1166, Nutriomics Team 6 (G.M., K.C.)
- Assistance Publique Hopitaux de Paris, AP-HP, Pitié-Salpêtrière Hospital, Nutrition and Endocrinology Department and Hepato-biliary and Digestive Surgery Department (K.C.)
| | - Sophie Hue
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- Sorbonne Université, INSERM UMRS 1166, Institute of Cardiometabolism and Nutrition ICAN (N.S., S.H.)
- AP-HP Vaccine Research Institute (VRI) (M.S., S.H.)
| | - Serge Adnot
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP, Department of Physiology, Henri Mondor Hospital, DHU-ATVB (S.A.)
| | - Stéphane N Hatem
- Institut de Cardiologie, Hôpital Universitaire Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (S.H.)
| | - Izuru Tsuchimochi
- Laboratory of Synthetic Organic and Medicinal Chemistry, Division of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University (I.T., T.Y.)
| | - Takehiko Yoshimitsu
- Laboratory of Synthetic Organic and Medicinal Chemistry, Division of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University (I.T., T.Y.)
| | - Corneliu Hénégar
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Geneviève Derumeaux
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP, Department of Cardiology, Henri Mondor Hospital, DHU-ATVB (J.T., G.D.)
| |
Collapse
|
34
|
Abstract
Cardiac ageing manifests as a decline in function leading to heart failure. At the cellular level, ageing entails decreased replicative capacity and dysregulation of cellular processes in myocardial and nonmyocyte cells. Various extrinsic parameters, such as lifestyle and environment, integrate important signalling pathways, such as those involving inflammation and oxidative stress, with intrinsic molecular mechanisms underlying resistance versus progression to cellular senescence. Mitigation of cardiac functional decline in an ageing organism requires the activation of enhanced maintenance and reparative capacity, thereby overcoming inherent endogenous limitations to retaining a youthful phenotype. Deciphering the molecular mechanisms underlying dysregulation of cellular function and renewal reveals potential interventional targets to attenuate degenerative processes at the cellular and systemic levels to improve quality of life for our ageing population. In this Review, we discuss the roles of extrinsic and intrinsic factors in cardiac ageing. Animal models of cardiac ageing are summarized, followed by an overview of the current and possible future treatments to mitigate the deleterious effects of cardiac ageing.
Collapse
|
35
|
Swirski FK, Nahrendorf M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol 2019; 18:733-744. [PMID: 30228378 DOI: 10.1038/s41577-018-0065-8] [Citation(s) in RCA: 488] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The past few decades have generated growing recognition that the immune system makes an important contribution to cardiac development, composition and function. Immune cells infiltrate the heart at gestation and remain in the myocardium, where they participate in essential housekeeping functions throughout life. After myocardial infarction or in response to infection, large numbers of immune cells are recruited to the heart to remove dying tissue, scavenge pathogens and promote healing. Under some circumstances, immune cells can cause irreversible damage, contributing to heart failure. This Review focuses on the role of the immune system in the heart under both homeostatic and perturbed conditions.
Collapse
Affiliation(s)
- Filip K Swirski
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
McShane L, Tabas I, Lemke G, Kurowska-Stolarska M, Maffia P. TAM receptors in cardiovascular disease. Cardiovasc Res 2019; 115:1286-1295. [PMID: 30980657 PMCID: PMC6587925 DOI: 10.1093/cvr/cvz100] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/28/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022] Open
Abstract
The TAM receptors are a distinct family of three receptor tyrosine kinases, namely Tyro3, Axl, and MerTK. Since their discovery in the early 1990s, they have been studied for their ability to influence numerous diseases, including cancer, chronic inflammatory and autoimmune disorders, and cardiovascular diseases. The TAM receptors demonstrate an ability to influence multiple aspects of cardiovascular pathology via their diverse effects on cells of both the vasculature and the immune system. In this review, we will explore the various functions of the TAM receptors and how they influence cardiovascular disease through regulation of vascular remodelling, efferocytosis and inflammation. Based on this information, we will suggest areas in which further research is required and identify potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Lucy McShane
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow, UK,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ira Tabas
- Departments of Medicine, Physiology, and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Greg Lemke
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA,Immunobiology and Microbial Pathogenesis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mariola Kurowska-Stolarska
- Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow, UK,Corresponding authors. Tel: +44 141 330 7142; E-mail: (P.M.) Tel: +44 141 330 6085; E-mail: (M.K.-S.)
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow, UK,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK,Department of Pharmacy, University of Naples Federico II, Naples, Italy,Corresponding authors. Tel: +44 141 330 7142; E-mail: (P.M.) Tel: +44 141 330 6085; E-mail: (M.K.-S.)
| |
Collapse
|
37
|
Humeres C, Frangogiannis NG. Fibroblasts in the Infarcted, Remodeling, and Failing Heart. JACC Basic Transl Sci 2019; 4:449-467. [PMID: 31312768 PMCID: PMC6610002 DOI: 10.1016/j.jacbts.2019.02.006] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
Expansion and activation of fibroblasts following cardiac injury is important for repair but may also contribute to fibrosis, remodeling, and dysfunction. The authors discuss the dynamic alterations of fibroblasts in failing and remodeling myocardium. Emerging concepts suggest that fibroblasts are not unidimensional cells that act exclusively by secreting extracellular matrix proteins, thus promoting fibrosis and diastolic dysfunction. In addition to their involvement in extracellular matrix expansion, activated fibroblasts may also exert protective actions, preserving the cardiac extracellular matrix, transducing survival signals to cardiomyocytes, and regulating inflammation and angiogenesis. The functional diversity of cardiac fibroblasts may reflect their phenotypic heterogeneity.
Collapse
Key Words
- AT1, angiotensin type 1
- ECM, extracellular matrix
- FAK, focal adhesion kinase
- FGF, fibroblast growth factor
- IL, interleukin
- MAPK, mitogen-activated protein kinase
- MRTF, myocardin-related transcription factor
- PDGF, platelet-derived growth factor
- RNA, ribonucleic acid
- ROCK, Rho-associated coiled-coil containing kinase
- ROS, reactive oxygen species
- SMA, smooth muscle actin
- TGF, transforming growth factor
- TRP, transient receptor potential
- cytokines
- extracellular matrix
- fibroblast
- infarction
- lncRNA, long noncoding ribonucleic acid
- miRNA, micro–ribonucleic acid
- remodeling
Collapse
Affiliation(s)
- Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
38
|
Hadzi‐Petrushev N, Angelovski M, Rebok K, Mitrokhin V, Kamkin A, Mladenov M. Antioxidant and anti‐inflammatory effects of the monocarbonyl curcumin analogs B2BRBC and C66 in monocrotaline‐induced right ventricular hypertrophy. J Biochem Mol Toxicol 2019; 33:e22353. [DOI: 10.1002/jbt.22353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 04/03/2019] [Accepted: 05/17/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Nikola Hadzi‐Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
| | - Marija Angelovski
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
| | - Katerina Rebok
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
| | - Vadim Mitrokhin
- Department of Fundamental and Applied PhysiologyRussian National Research Medical University Moscow Russia
| | - Andre Kamkin
- Department of Fundamental and Applied PhysiologyRussian National Research Medical University Moscow Russia
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
- Department of Fundamental and Applied PhysiologyRussian National Research Medical University Moscow Russia
| |
Collapse
|
39
|
Nagel F, Santer D, Stojkovic S, Kaun C, Schaefer AK, Krššák M, Abraham D, Bencsik P, Ferdinandy P, Kenyeres E, Szabados T, Wojta J, Trescher K, Kiss A, Podesser BK. The impact of age on cardiac function and extracellular matrix component expression in adverse post-infarction remodeling in mice. Exp Gerontol 2019; 119:193-202. [PMID: 30763602 DOI: 10.1016/j.exger.2019.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 01/18/2023]
Abstract
The aim of this study was to describe the potential associations of the expression of matricellular components in adverse post-infarction remodeling of the geriatric heart. In male geriatric (OM, age: 18 months) and young (YM, age: 11 weeks) OF1 mice myocardial infarction (MI) was induced by permanent ligation of the left anterior descending coronary artery. Cardiac function was evaluated by MRI. Plasma and myocardial tissue samples were collected 3d, 7d, and 32d post-MI. Age and MI were associated with impaired cardiac function accompanied by left-ventricular (LV) dilatation. mRNA expression of MMP-2 (7d: p < 0.05), TIMP-1 (7d: p < 0.05), TIMP-2 (7d: p < 0.05), Collagen-1 (3d and 7d: p < 0.05) and Collagen-3 (7d: p < 0.05) in LV non-infarcted myocardium was significantly higher in YM than in OM after MI. MMP-9 activity in plasma was increased in OM after MI (3d: p < 0.01). Tenascin-C protein levels assessed by ELISA were decreased in OM as compared to YM after MI in plasma (3d: p < 0.001, 7d: p < 0.05) and LV non-infarcted myocardium (7d: p < 0.01). Dysregulation in ECM components in non-infarcted LV might be associated and contribute to adverse LV remodeling and impaired cardiac function. Thus, targeting ECM might be a potential therapeutic approach to enhance cardiac function in geriatric patients following MI.
Collapse
Affiliation(s)
- Felix Nagel
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiac Surgery, University Hospital St. Poelten, Dunant-Platz 1, 3100 St. Poelten, Austria
| | - David Santer
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiovascular Surgery, Hospital Hietzing, Wolkersbergenstr. 1, 1130 Wien, Austria
| | - Stefan Stojkovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria
| | - Christoph Kaun
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria
| | - Anne-Kristin Schaefer
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria
| | - Martin Krššák
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria; High Field MR Centre, Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, Lazarettg. 14, 1090 Wien, Austria
| | - Dietmar Abraham
- Laboratory for Molecular Cellular Biology, Medical University of Vienna, Schwarzspanierstr. 17, 1090 Wien, Austria
| | - Péter Bencsik
- Pharmahungary Group, Szeged, Hungary; Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dom ter 12, 6721 Szeged, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, Budapest 1089, Hungary
| | - Eva Kenyeres
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dom ter 12, 6721 Szeged, Hungary
| | - Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dom ter 12, 6721 Szeged, Hungary
| | - Johann Wojta
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria
| | - Karola Trescher
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiac Surgery, University Hospital St. Poelten, Dunant-Platz 1, 3100 St. Poelten, Austria
| | - Attila Kiss
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiac Surgery, University Hospital St. Poelten, Dunant-Platz 1, 3100 St. Poelten, Austria.
| |
Collapse
|
40
|
de Boer RA, De Keulenaer G, Bauersachs J, Brutsaert D, Cleland JG, Diez J, Du XJ, Ford P, Heinzel FR, Lipson KE, McDonagh T, Lopez-Andres N, Lunde IG, Lyon AR, Pollesello P, Prasad SK, Tocchetti CG, Mayr M, Sluijter JPG, Thum T, Tschöpe C, Zannad F, Zimmermann WH, Ruschitzka F, Filippatos G, Lindsey ML, Maack C, Heymans S. Towards better definition, quantification and treatment of fibrosis in heart failure. A scientific roadmap by the Committee of Translational Research of the Heart Failure Association (HFA) of the European Society of Cardiology. Eur J Heart Fail 2019; 21:272-285. [PMID: 30714667 PMCID: PMC6607480 DOI: 10.1002/ejhf.1406] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022] Open
Abstract
Fibrosis is a pivotal player in heart failure development and progression. Measurements of (markers of) fibrosis in tissue and blood may help to diagnose and risk stratify patients with heart failure, and its treatment may be effective in preventing heart failure and its progression. A lack of pathophysiological insights and uniform definitions has hampered the research in fibrosis and heart failure. The Translational Research Committee of the Heart Failure Association discussed several aspects of fibrosis in their workshop. Early insidious perturbations such as subclinical hypertension or inflammation may trigger first fibrotic events, while more dramatic triggers such as myocardial infarction and myocarditis give rise to full blown scar formation and ongoing fibrosis in diseased hearts. Aging itself is also associated with a cardiac phenotype that includes fibrosis. Fibrosis is an extremely heterogeneous phenomenon, as several stages of the fibrotic process exist, each with different fibrosis subtypes and a different composition of various cells and proteins — resulting in a very complex pathophysiology. As a result, detection of fibrosis, e.g. using current cardiac imaging modalities or plasma biomarkers, will detect only specific subforms of fibrosis, but cannot capture all aspects of the complex fibrotic process. Furthermore, several anti‐fibrotic therapies are under investigation, but such therapies generally target aspecific aspects of the fibrotic process and suffer from a lack of precision. This review discusses the mechanisms and the caveats and proposes a roadmap for future research.
Collapse
Affiliation(s)
- Rudolf A de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, Groningen, The Netherlands
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Dirk Brutsaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - John G Cleland
- Robertson Centre for Biostatistics & Clinical Trials, University of Glasgow, Glasgow, UK
| | - Javier Diez
- Program of Cardiovascular Diseases, Center for Applied Medical Research, Departments of Nephrology, and Cardiology and Cardiac Surgery, University Clinic, University of Navarra, Pamplona, Spain
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | | | - Frank R Heinzel
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | | | | | - Natalia Lopez-Andres
- Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Publica de Navarra, Idisna, Spain
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Alexander R Lyon
- Royal Brompton Hospital, and Imperial College London, London, UK
| | | | | | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Manuel Mayr
- The James Black Centre, King's College, University of London, London, UK
| | - Joost P G Sluijter
- University Medical Centre Utrecht, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Center, University Utrecht, Utrecht, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany.,DZHK (German Center for Cardiovascular Research) partner site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | - Faiez Zannad
- Centre d'Investigation Clinique, CHU de Nancy, Nancy, France
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research) partner site Göttingen, Göttingen, Germany
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Gerasimos Filippatos
- Heart Failure Unit, Department of Cardiology, School of Medicine, Athens University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
| | - Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | - Christoph Maack
- Comprehensive Heart Failure Centre, University and University Hospital Würzburg, Würzburg, Germany
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium.,The Netherlands Heart Institute, Nl-HI, Utrecht, The Netherlands
| |
Collapse
|
41
|
Leoni G, Soehnlein O. (Re) Solving Repair After Myocardial Infarction. Front Pharmacol 2018; 9:1342. [PMID: 30534069 PMCID: PMC6275178 DOI: 10.3389/fphar.2018.01342] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/31/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases, including myocardial infarction and its complications such as heart failure, are the leading cause of death worldwide. To date, basic and translational research becomes necessary to unravel the mechanisms of cardiac repair post-myocardial infarction. The local inflammatory tissue response after acute myocardial infarction determines the subsequent healing process. The diversity of leukocytes such as neutrophils, macrophages and lymphocytes contribute to the clearance of dead cells while activating reparative pathways necessary for myocardial healing. Cardiomyocyte death triggers wall thinning, ventricular dilatation, and fibrosis that can cause left ventricular dysfunction and heart failure. The ultimate goal of cardiac repair is to regenerate functionally viable myocardium after myocardial infarction to prevent cardiac death. Current therapies for heart failure after myocardial infarction are limited and non-curative. At the moment in clinic, conventional surgical interventions such as coronary artery bypass graft or percutaneous coronary interventions are only able to partially restore heart function, with a minor improvement in the left ventricular ejection fraction. The goal of this review is to provide an overview of endogenous myocardial repair mechanisms possibly transferable to future treatment strategies. Among the innovative factors identified as essential in cardiac healing, we highlight specialized pro-resolving mediators as the emerging factors that provide the key molecular signals for the activation of the reparative cells in the myocardium.
Collapse
Affiliation(s)
- Giovanna Leoni
- Institute for Cardiovascular Prevention (IPEK), University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Physiology and Pharmacology (FyFa), Karolinska Institute, Stockholm, Sweden.,Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
42
|
Stoyek MR, Rog-Zielinska EA, Quinn TA. Age-associated changes in electrical function of the zebrafish heart. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:91-104. [DOI: 10.1016/j.pbiomolbio.2018.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 12/17/2022]
|
43
|
Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med 2018; 65:70-99. [PMID: 30056242 DOI: 10.1016/j.mam.2018.07.001] [Citation(s) in RCA: 533] [Impact Index Per Article: 76.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic companion of most myocardial diseases, and is associated with systolic and diastolic dysfunction, arrhythmogenesis, and adverse outcome. Because the adult mammalian heart has negligible regenerative capacity, death of a large number of cardiomyocytes results in reparative fibrosis, a process that is critical for preservation of the structural integrity of the infarcted ventricle. On the other hand, pathophysiologic stimuli, such as pressure overload, volume overload, metabolic dysfunction, and aging may cause interstitial and perivascular fibrosis in the absence of infarction. Activated myofibroblasts are the main effector cells in cardiac fibrosis; their expansion following myocardial injury is primarily driven through activation of resident interstitial cell populations. Several other cell types, including cardiomyocytes, endothelial cells, pericytes, macrophages, lymphocytes and mast cells may contribute to the fibrotic process, by producing proteases that participate in matrix metabolism, by secreting fibrogenic mediators and matricellular proteins, or by exerting contact-dependent actions on fibroblast phenotype. The mechanisms of induction of fibrogenic signals are dependent on the type of primary myocardial injury. Activation of neurohumoral pathways stimulates fibroblasts both directly, and through effects on immune cell populations. Cytokines and growth factors, such as Tumor Necrosis Factor-α, Interleukin (IL)-1, IL-10, chemokines, members of the Transforming Growth Factor-β family, IL-11, and Platelet-Derived Growth Factors are secreted in the cardiac interstitium and play distinct roles in activating specific aspects of the fibrotic response. Secreted fibrogenic mediators and matricellular proteins bind to cell surface receptors in fibroblasts, such as cytokine receptors, integrins, syndecans and CD44, and transduce intracellular signaling cascades that regulate genes involved in synthesis, processing and metabolism of the extracellular matrix. Endogenous pathways involved in negative regulation of fibrosis are critical for cardiac repair and may protect the myocardium from excessive fibrogenic responses. Due to the reparative nature of many forms of cardiac fibrosis, targeting fibrotic remodeling following myocardial injury poses major challenges. Development of effective therapies will require careful dissection of the cell biological mechanisms, study of the functional consequences of fibrotic changes on the myocardium, and identification of heart failure patient subsets with overactive fibrotic responses.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B, Bronx, NY, 10461, USA.
| |
Collapse
|
44
|
Wang X, Fan X, Ji S, Ma A, Wang T. Prognostic value of neutrophil to lymphocyte ratio in heart failure patients. Clin Chim Acta 2018; 485:44-49. [PMID: 29932879 DOI: 10.1016/j.cca.2018.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/27/2018] [Accepted: 06/14/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Neutrophil to lymphocyte ratio (NLR) has been indicated to be an independent predictor for all-cause mortality or adverse events in a variety of diseases. However, no consistent conclusions regarding it's relevance to patients with heart failure have been made. This meta-analysis was conducted to assess the significance of NLR in patients with heart failure. METHODS Pubmed and Embase databases were searched for eligible studies that reported the association between NLR and heart failure through September 2017. The overall hazard ratio (HR) and corresponding 95% confidence interval (CI) were used to assess the associations. RESULTS Ten studies met the eligibility criteria and a total of 5979 heart failure patients were included in the meta-analysis. The overall hazard ratio (HR) for all-cause mortality was 1.28 (95% CI 1.14-1.43) and the HR of renal dysfunction was 1.23 (95% CI 1.07-1.41) comparing the highest with the lowest category of NLR. However, the total pooled adjusted HR for in-hospital mortality (HR = 1.18, 95% CI 0.88-1.59) and rehospitalization (HR = 2.19, 95% CI 0.94-5.09) were not statistically significant. A subgroup analysis showed that sample size with moderate heterogeneity may be the origin of heterogeneity in all-cause mortality. Sensitivity analysis proved the stability of results of our meta-analysis. CONCLUSIONS The meta-analysis demonstrates that NLR is a predictor of all-cause mortality in patients with heart failure. Because the quality of the included studies varies, further well-designed studies are needed to confirm this association.
Collapse
Affiliation(s)
- Xiqiang Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China
| | - Xiude Fan
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China
| | - Shuaifei Ji
- Department of Ophthalmology, TangDu Hospital, The Fourth Military University, Xi'an 710032, Shaanxi, PR China
| | - Aiqun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China; Key Laboratory of Molecular Cardiology, Shaanxi Province, PR China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, PR China.
| | - Tingzhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China; Key Laboratory of Molecular Cardiology, Shaanxi Province, PR China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, PR China.
| |
Collapse
|
45
|
Shaikh N, Sardar M, Jacob A, Alagusundaramoorthy SS, Eng M, Checton J, Shah A. Possible predictive factors for recovery of left ventricular systolic function in Takotsubo cardiomyopathy. Intractable Rare Dis Res 2018; 7:100-105. [PMID: 29862151 PMCID: PMC5982616 DOI: 10.5582/irdr.2018.01042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Takotsubo cardiomyopathy (TTC) is a transient systolic dysfunction of the left ventricle which is usually seen in elderly women, often following a physical or emotional stressful event. Little is known about the prognostic factors affecting the recovery of systolic function. Thirty-six patients diagnosed with TTC from January 2006 to January 2017 at our hospital were included. Median time to recovery of ejection fraction (EF) was calculated to be 25 days. Early recovery of ejection fraction was defined as less than or equal to 25 days (group 1) and late recovery was defined as more than 25 days (group 2). Demographic and clinical factors were compared between the groups. Fifty percent patients had early recovery of EF with a mean time to recovery of 7.11 days and 50% had late recovery of ejection fraction with a mean time to recovery of 58.38 days. Younger age at presentation was associated with early recovery of systolic function (58.83 ± 2.7 years vs. 67.33 ± 2.7 years, p = 0 .032). Presence of an identifiable triggering event was associated with early recovery (83% in group 1 vs. 50% in group 2, p = 0.034). Generalized anxiety disorder was seen more commonly in the group with early recovery (78% in group 1 vs. 45% in group 2, p = 0.040). In conclusion, younger age, generalized anxiety disorder and presence of triggering event were seen more commonly in patients with early recovery of left ventricular systolic function in Takotsubo cardiomyopathy.
Collapse
Affiliation(s)
- Nasreen Shaikh
- Department of Medicine, Monmouth Medical Center, Long Branch, NJ, USA
- Address correspondence to:Dr. Nasreen Shaikh, Department of Medicine, Monmouth Medical Center, 300, Second Avenue, Long Branch, NJ 07740, USA. E-mail:
| | - Muhammad Sardar
- Department of Medicine, Monmouth Medical Center, Long Branch, NJ, USA
| | - Aasems Jacob
- Department of Medicine, Monmouth Medical Center, Long Branch, NJ, USA
| | | | - Margaret Eng
- Department of Medicine, Monmouth Medical Center, Long Branch, NJ, USA
| | - John Checton
- Department of Medicine, Monmouth Medical Center, Long Branch, NJ, USA
| | - Ajay Shah
- Department of Medicine, Monmouth Medical Center, Long Branch, NJ, USA
| |
Collapse
|
46
|
Rizzo P, Bollini S, Bertero E, Ferrari R, Ameri P. Beyond cardiomyocyte loss: Role of Notch in cardiac aging. J Cell Physiol 2018; 233:5670-5683. [PMID: 29271542 DOI: 10.1002/jcp.26417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022]
Abstract
The knowledge of the cellular events occurring in the aging heart has dramatically expanded in the last decade and is expected to further grow in years to come. It is now clear that impaired function and loss of cardiomyocytes are major features of cardiac aging, but other events are likewise important. In particular, accumulating experimental evidence highlights the importance of fibroblast and cardiac progenitor cell (CPC) dysfunction. The Notch pathway regulates cardiomyocyte, fibroblast, and CPC activity and, thus, may be critically involved in heart disease associated with advanced age, especially heart failure. In a translational perspective, thorough investigation of the Notch system in the aging myocardium may lead to the identification of molecular targets for novel therapies for age-related cardiac disease.
Collapse
Affiliation(s)
- Paola Rizzo
- Department of Morphology, Surgery, and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care and Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Sveva Bollini
- Department of Experimental Medicine, Regenerative Medicine Laboratory, University of Genova, Genova, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, Laboratory of Cardiovascular Biology, University of Genova and Ospedale Policlinico San Martino IRCCS per Oncologia, Genova, Italy
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care and Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Pietro Ameri
- Department of Internal Medicine, Laboratory of Cardiovascular Biology, University of Genova and Ospedale Policlinico San Martino IRCCS per Oncologia, Genova, Italy
| |
Collapse
|
47
|
Boman K, Thormark Fröst F, Bergman ACR, Olofsson M. NTproBNP and ST2 as predictors for all-cause and cardiovascular mortality in elderly patients with symptoms suggestive for heart failure. Biomarkers 2018; 23:373-379. [PMID: 29355441 DOI: 10.1080/1354750x.2018.1431692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND A new biomarker, suppression of tumorigenicity 2 (ST2) has been introduced as a marker for fibrosis and hypertrophy. Its clinical value in comparison with N-terminal pro-hormone of brain natriuretic peptide /Amino-terminal pro-B-type natriuretic peptide (NTproBNP) in predicting mortality in elderly patients with symptoms of heart failure (HF) is still unclear. AIM To evaluate the prognostic value for all-cause- and cardiovascular mortality of ST2 or NTproBNP and the combination of these biomarkers. PATIENTS AND METHODS One hundred seventy patients patients with clinical symptoms of HF (77 (45%) were with verified HF) were recruited from one selected primary health care center (PHC) in Sweden and echocardiography was performed in all patients. Blood samples were obtained from 159 patients and stored frozen at -70 °C. NTproBNP was analyzed at a central core laboratory using a clinically available immunoassay.ST2 was analyzed with Critical Diagnostics Presage ST2 ELISA immunoassay. RESULTS We studied 159 patients (mean age 77 ± 8.3 years, 70% women). During ten years of follow up 78 patients had died, out of which 50 deaths were for cardiovascular reasons. Continuous NTproBNP and ST2 were both significantly associated with all-cause mortality (1.0001; 1.00001-1.0002, p = 0.04 and 1.03; 1.003-1.06, p = 0.03), NTproBNP but not ST2 remained significant for cardiovascular mortality after adjustments (1.0001; 1.00001-1.0002, p = 0.03 and 1.01; 0.77-1.06, p = 0.53), respectively. NTproBNP above median (>328 ng/L) compared to below median was significantly associated with all-cause mortality(HR: 4.0; CI :2.46-6.61; p < 0.001) and cardiovascular mortality (HR: 6.1; CI: 3.11-11.95; p < 0.001). Corresponding analysis for ST2 above median (25.6 ng/L) was not significantly associated neither with all-cause mortality (HR; 1.4; CI: 0.89-2.77) nor cardiovascular mortality (HR: 1.3; CI: 0.73-2.23) and no significant interaction of NTproBNP and ST2 (OR: 1.1; CI: 0.42-3.12) was found. CONCLUSION In elderly patients with symptoms of heart failure ST2 was not superior to NTproBNP to predict all cause or cardiovascular mortality. Furthermore, it is unclear if the combination of ST2 and NTproBNP will improve long-term prognostication beyond what is achieved by NTproBNP alone.
Collapse
Affiliation(s)
- Kurt Boman
- a Research Unit, Department of Medicine , Skellefteå Hospital , Skellefteå , Sweden.,b Department of Public Health and Clinical Medicine , Umeå University , Umeå , Sweden
| | - Finn Thormark Fröst
- c Department of Clinical Chemistry , Karolinska University Hospital , Stockholm , Sweden
| | | | - Mona Olofsson
- a Research Unit, Department of Medicine , Skellefteå Hospital , Skellefteå , Sweden
| |
Collapse
|
48
|
Li L, Zhao Q, Kong W. Extracellular matrix remodeling and cardiac fibrosis. Matrix Biol 2018; 68-69:490-506. [PMID: 29371055 DOI: 10.1016/j.matbio.2018.01.013] [Citation(s) in RCA: 242] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022]
Abstract
Cardiac fibrosis, characterized by excessive deposition of extracellular matrix (ECM) proteins in the myocardium, distorts the architecture of the myocardium, facilitates the progression of arrhythmia and cardiac dysfunction, and influences the clinical course and outcome in patients with heart failure. This review describes the composition and homeostasis in normal cardiac interstitial matrix and introduces cellular and molecular mechanisms involved in cardiac fibrosis. We also characterize the ECM alteration in the fibrotic response under diverse cardiac pathological conditions and depict the role of matricellular proteins in the pathogenesis of cardiac fibrosis. Moreover, the diagnosis of cardiac fibrosis based on imaging and biomarker detection and the therapeutic strategies are addressed. Understanding the comprehensive molecules and pathways involved in ECM homeostasis and remodeling may provide important novel potential targets for preventing and treating cardiac fibrosis.
Collapse
Affiliation(s)
- Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Qian Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
49
|
Egemnazarov B, Crnkovic S, Nagy BM, Olschewski H, Kwapiszewska G. Right ventricular fibrosis and dysfunction: Actual concepts and common misconceptions. Matrix Biol 2018; 68-69:507-521. [PMID: 29343458 DOI: 10.1016/j.matbio.2018.01.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/25/2022]
Abstract
Fibrosis and remodeling of the right ventricle (RV) are associated with RV dysfunction and mortality of patients with pulmonary hypertension (PH) but it is unknown how much RV fibrosis contributes to RV dysfunction and mortality. RV fibrosis manifests as fibroblast accumulation and collagen deposition which may be excessive. Although extracellular matrix deposition leads to elevated ventricular stiffness, it is not known to which extent it affects RV function. Various animal models of pulmonary hypertension have been established to investigate the role of fibrosis in RV dysfunction and failure. However, they do not perfectly resemble the human disease. In the current review we describe the major characteristics of RV fibrosis, molecular mechanisms regulating the fibrotic process, and discuss how therapeutic targeting of fibrosis might affect RV function.
Collapse
Affiliation(s)
| | - Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Bence M Nagy
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Institute of Physiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
50
|
Sibiya N, Ngubane P, Mabandla M. Cardioprotective effects of pectin-insulin patch in streptozotocin-induced diabetic rats. J Diabetes 2017; 9:1073-1081. [PMID: 28220624 DOI: 10.1111/1753-0407.12538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/16/2017] [Accepted: 02/14/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Cardiovascular complications are among the leading causes of morbidity and mortality in diabetes mellitus. Despite the beneficial effects of subcutaneous insulin, reports suggest that the therapy itself precipitates cardiovascular risks due to the high insulin concentration administered. It is therefore necessary to seek alternative routes of insulin administration that may bypass the undesirable effects associated with high plasma insulin concentrations. Accordingly, the present study investigated the effects of a novel transdermal pectin-insulin patch on selected markers of cardiovascular function in diabetes. METHODS Pectin-insulin matrix patches (20.0, 40.8, and 82.9 μg/kg) were prepared as described previously. The three formulations were applied to streptozotocin-induced diabetic rats thrice daily. Blood glucose concentrations and mean arterial pressure (MAP) were monitored weekly for 5 weeks. Rats were then killed and blood collected for analysis of the lipid profile, cardiotropin-1, tumor necrosis factor (TNF)-α, and high-sensitivity C-reactive protein (hsCRP). RESULTS The patches decreased blood glucose concentrations and diabetes-induced disturbances in lipid profile were attenuated by patch application (82.9 μg/kg). The diabetes-induced increase in MAP was also attenuated in patch (82.9 μg/kg)-treated rats. Patch treatment resulted in a decreased heart weight: body weight ratio, as well as reductions in cardiotropin-1, TNF-α, and hsCRP concentrations. CONCLUSIONS Application of the pectin-insulin patch protects against the debilitating cardiovascular effects associated with conventional diabetes treatment. This suggests that the pectin-insulin patch may provide an effective alternative therapeutic approach to the commonly used subcutaneous insulin injections in the management of diabetes.
Collapse
Affiliation(s)
- Ntethelelo Sibiya
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Phikelelani Ngubane
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Musa Mabandla
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|