1
|
Guo X, Wang P, Wei H, Yan J, Zhang D, Qian Y, Guo B. Interleukin(IL)-37 attenuates isoproterenol (ISO)-induced cardiac hypertrophy by suppressing JAK2/STAT3-signaling associated inflammation and oxidative stress. Int Immunopharmacol 2024; 142:113134. [PMID: 39293311 DOI: 10.1016/j.intimp.2024.113134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Inflammation and oxidative stress have drawn more and more interest in the realm of cardiovascular disease. In many different disorders, IL-37 acts as an anti-inflammatory and suppressor of inflammation. This study aimed to investigate whether IL-37 could alleviate cardiac hypertrophy by reducing inflammation and oxidative stress. METHODS In vivo, a cardiac hypertrophy model was induced by 14 d of daily isoproterenol (ISO, 30 mg/kg/d) injection, followed by weeks of treatment with recombinant human IL-37 (1000 ng/animal), administered three times weekly. Assessments concentrated on markers of inflammation and oxidative stress, apoptosis, myocardial disease, and cardiac shape and function. In vitro, neonatal rat cardiomyocytes (NRCMs) were subjected to ISO (10 µM) to establish a cardiomyocytes hypertrophy model. Subsequent IL-37 treatment (100 ng/ml) was applied to determine its cardioprotective efficacy and to elucidate further the underlying mechanisms involved. RESULTS Significant cardioprotective benefits of IL-37 were seen (in vitro as well as in vivo), primarily through the reduction of oxidative stress, inflammation, apoptosis, and heart hypertrophy markers. Furthermore, IL-37 treatment was associated with a decrease in JAK2 and STAT3 phosphorylation. It is interesting to note that WP1066, a JAK2/STAT3 inhibitor, exhibited antioxidant and anti-inflammatory properties comparable to IL-37, as well as synergistic effects when mixed with the latter. CONCLUSION ISO-induced cardiac hypertrophy is lessened by IL-37 through the reduction of oxidative stress and inflammation. Additionally, the effects of IL-37 are closely related to inactivation of the JAK2/STAT3 signaling pathway. It is anticipated that IL-37 will one day be used to treat cardiovascular illnesses such as heart hypertrophy.
Collapse
Affiliation(s)
- Xiaohua Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Pengfei Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Huiqing Wei
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Jie Yan
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Donglei Zhang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Yuxing Qian
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China; Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
2
|
Shahsavarnajand Bonab H, Tolouei Azar J, Soraya H, Nouri Habashi A. Aerobic interval training preconditioning protocols inhibit isoproterenol-induced pathological cardiac remodeling in rats: Implications on oxidative balance, autophagy, and apoptosis. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:344-357. [PMID: 39309465 PMCID: PMC11411311 DOI: 10.1016/j.smhs.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 09/25/2024] Open
Abstract
This study aimed to investigate the potential cardioprotective effects of moderate and high-intensity aerobic interval training (MIIT and HIIT) preconditioning. The focus was on histological changes, pro-oxidant-antioxidant balance, autophagy initiation, and apoptosis in myocardial tissue incited by isoproterenol-induced pathological cardiac remodeling (ISO-induced PCR). Male Wistar rats were randomly divided into control (n = 6), ISO (n = 8), MIIT (n = 4), HIIT (n = 4), MIIT + ISO (n = 8), and HIIT + ISO (n = 8) groups. The MIIT and HIIT protocols were administered for 10 weeks, followed by the induction of cardiac remodeling using subcutaneous injection of ISO (100 mg/kg for two consecutive days). Alterations in heart rate (HR), mean arterial pressure (MAP), rate pressure product (RPP), myocardial oxygen consumption (MV ˙ O2), cardiac hypertrophy, histopathological changes, pro-oxidant-antioxidant balance, autophagy biomarkers (Beclin-1, Atg7, p62, LC3 I/II), and apoptotic cell distribution were measured. The findings revealed that the MIIT + ISO and HIIT + ISO groups demonstrated diminished myocardial damage, hemorrhage, immune cell infiltration, edema, necrosis, and apoptosis compared to ISO-induced rats. MIIT and HIIT preconditioning mitigated HR, enhanced MAP, and preserved MV ˙ O2 and RPP. The pro-oxidant-antioxidant balance was sustained in both MIIT + ISO and HIIT + ISO groups, with MIIT primarily inhibiting pro-apoptotic autophagy progression through maintaining pro-oxidant-antioxidant balance, and HIIT promoting pro-survival autophagy. The results demonstrated the beneficial effects of both MIIT and HIIT as AITs preconditioning in ameliorating ISO-induced PCR by improving exercise capacity, hemodynamic parameters, and histopathological changes. Some of these protective effects can be attributed to the modulation of cardiac apoptosis, autophagy, and oxidative stress.
Collapse
Affiliation(s)
- Hakimeh Shahsavarnajand Bonab
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Javad Tolouei Azar
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Akbar Nouri Habashi
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| |
Collapse
|
3
|
Anilkumar S A, Dutta S, Aboo S, Ismail A. Vitamin D as a modulator of molecular pathways involved in CVDs: Evidence from preclinical studies. Life Sci 2024; 357:123062. [PMID: 39288869 DOI: 10.1016/j.lfs.2024.123062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/27/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Vitamin D deficiency (VDD) is a widespread global health issue, affecting nearly a billion individuals worldwide, and mounting evidence links it to an increased risk of cardiovascular diseases like hypertension, atherosclerosis, and heart failure. The discovery of vitamin D receptors and metabolizing enzymes in cardiac and vascular cells, coupled with experimental studies, underscores the complex relationship between vitamin D and cardiovascular health. This review aims to synthesize and critically evaluate the preclinical evidence elucidating the role of vitamin D in cardiovascular health. We examined diverse preclinical in vitro (cardiomyocyte cell line) models and in vivo models, including knockout mice, diet-induced deficiency, and disease-specific animal models (hypertension, hypertrophy and myocardial infarction). These studies reveal that vitamin D modulates vascular tone, and prevents fibrosis and hypertrophy through effects on major signal transduction pathways (NF-kB, Nrf2, PI3K/AKT/mTOR, Calcineurin/NFAT, TGF-β/Smad, AMPK) and influences epigenetic mechanisms governing inflammation, oxidative stress, and pathological remodeling. In vitro studies elucidate vitamin D's capacity to promote cardiomyocyte differentiation and inhibit pathological remodeling. In vivo studies further uncovered detrimental cardiac effects of VDD, while supplementation with vitamin D in cardiovascular disease (CVD) models demonstrated its protective effects by decreasing inflammation, attenuating hypertrophy, reduction in plaque formation, and improving cardiac function. Hence, this comprehensive review emphasizes the critical role of vitamin D in cardiovascular health and its potential as a preventive/therapeutic strategy in CVDs. However, further research is needed to translate these findings into clinical applications as there are discrepancies between preclinical and clinical studies.
Collapse
Affiliation(s)
- Athira Anilkumar S
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Soumam Dutta
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Shabna Aboo
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India.
| | - Ayesha Ismail
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
4
|
Choudhury P, Kandula N, Kosuru R, Adena SKR. Nanomedicine: A great boon for cardiac regenerative medicine. Eur J Pharmacol 2024; 982:176969. [PMID: 39218342 DOI: 10.1016/j.ejphar.2024.176969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cardiovascular disease (CVD) represents a significant global health challenge, remaining the leading cause of illness and mortality worldwide. The adult heart's limited regenerative capacity poses a major obstacle in repairing extensive damage caused by conditions like myocardial infarction. In response to these challenges, nanomedicine has emerged as a promising field aimed at improving treatment outcomes through innovative drug delivery strategies. Nanocarriers, such as nanoparticles (NPs), offer a revolutionary approach by facilitating targeted delivery of therapeutic agents directly to the heart. This precise delivery system holds immense potential for treating various cardiac conditions by addressing underlying mechanisms such as inflammation, oxidative stress, cell death, extracellular matrix remodeling, prosurvival signaling, and angiogenic pathways associated with ischemia-reperfusion injury. In this review, we provide a concise summary of the fundamental mechanisms involved in cardiac remodeling and regeneration. We explore how nanoparticle-based drug delivery systems can effectively target the afore-mentioned mechanisms. Furthermore, we discuss clinical trials that have utilized nanoparticle-based drug delivery systems specifically designed for cardiac applications. These trials demonstrate the potential of nanomedicine in clinical settings, paving the way for future advancements in cardiac therapeutics through precise and efficient drug delivery. Overall, nanomedicine holds promise in revolutionizing the treatment landscape of cardiovascular diseases by offering targeted and effective therapeutic strategies that address the complex pathophysiology of cardiac injuries.
Collapse
Affiliation(s)
- Priyanka Choudhury
- Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Nirupama Kandula
- Department of Microbiology, GSL Medical College, Rajahmahendravaram, Andhra Pradesh, 533296, India
| | - Ramoji Kosuru
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
| | - Sandeep Kumar Reddy Adena
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
5
|
Athmuri DN, Bhattacharyya J, Bhatnagar N, Shiekh PA. Alleviating hypoxia and oxidative stress for treatment of cardiovascular diseases: a biomaterials perspective. J Mater Chem B 2024; 12:10490-10515. [PMID: 39302443 DOI: 10.1039/d4tb01126k] [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: 09/22/2024]
Abstract
A state of hypoxia (lack of oxygen) persists in the initial and later phases of healing in cardiovascular diseases, which can alter the tissue's repair or regeneration, ultimately affecting the structure and functionality of the related organ. Consequently, this results in a cascade of events, leading to metabolic stress and the production of reactive oxygen species (ROS) and autophagy. This unwanted situation not only limits the oxygen supply to the needy tissues but also creates an inflammatory state, limiting the exchange of nutrients and other supplements. Consequently, biomaterials have gained considerable attention to alleviate hypoxia and oxidative stress in cardiovascular diseases. Numerous oxygen releasing and antioxidant biomaterials have been developed and proven to alleviate hypoxia and oxidative stress. This review article summarizes the mechanisms involved in cardiovascular pathologies due to hypoxia and oxidative stress, as well as the treatment modalities currently in practice. The applications, benefits and possible shortcomings of these approaches have been discussed. Additionally, the review explores the role of novel biomaterials in combating the limitations of existing approaches, primarily focusing on the development of oxygen-releasing and antioxidant biomaterials for cardiac repair and regeneration. It also directs attention to various other potential applications with critical insights for further advancement in this area.
Collapse
Affiliation(s)
- Durga Nandini Athmuri
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| | - Jayanta Bhattacharyya
- Bio-therapeutics Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India
| | - Naresh Bhatnagar
- Department of Mechanical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India
| | - Parvaiz Ahmad Shiekh
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| |
Collapse
|
6
|
Pena E, El Alam S, Gonzalez C, Cortés I, Aguilera D, Flores K, Arriaza K. Astaxanthin Supplementation Effects in Right Ventricle of Rats Exposed to Chronic Intermittent Hypobaric Hypoxia. Antioxidants (Basel) 2024; 13:1269. [PMID: 39456521 PMCID: PMC11504862 DOI: 10.3390/antiox13101269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
In Chile, individuals are commonly exposed to high altitude due to the work shift system, involving days of exposure to high altitude followed by days at sea level over the long term, which can result in chronic intermittent hypobaric hypoxia (CIHH). CIHH can cause high-altitude pulmonary hypertension (HAPH), the principal manifestation of which is right ventricular hypertrophy (RVH), in some cases leading to heart failure and eventually death. Studies have shown the contribution of oxidative stress and inflammation to RVH development. Recently, it was determined that the pigment astaxanthin has high antioxidant capacity and strong anti-inflammatory and cardioprotective effects. Therefore, the aim of this study was to determine the effects of astaxanthin on RVH development in rats subjected to CIHH. METHODS Thirty two male Wistar rats were randomly assigned to the following groups (n = 8 per group): the normoxia with vehicle (NX), normoxia with astaxanthin (NX + AS), chronic intermittent hypobaric hypoxia with vehicle (CIHH), and chronic intermittent hypobaric hypoxia with astaxanthin (CIHH + AS) groups. CIHH was simulated by 2 days in a hypobaric chamber followed by 2 days at sea level for 29 days. RESULTS Exposure to CIHH induced RVH and increased lipid peroxidation (MDA), Nox2 expression, and SOD activity, however, it decreased pro-IL-1β expression. Astaxanthin restored oxidative stress markers (Nox2 and MDA), increased GPx activity, and decreased RVH compared to CIHH. CONCLUSION Astaxanthin alleviates RVH and reduces Nox2 and MDA levels while increasing GPx activity in rats subjected to CIHH. These findings provide new insights of astaxanthin as a new nutraceutical against high-altitude effects.
Collapse
Affiliation(s)
- Eduardo Pena
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1100000, Chile; (S.E.A.); (C.G.); (D.A.); (K.F.); (K.A.)
| | - Samia El Alam
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1100000, Chile; (S.E.A.); (C.G.); (D.A.); (K.F.); (K.A.)
| | - Constanza Gonzalez
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1100000, Chile; (S.E.A.); (C.G.); (D.A.); (K.F.); (K.A.)
| | - Isaac Cortés
- Science Faculty, Arturo Prat University, Iquique 1100000, Chile;
| | - Diego Aguilera
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1100000, Chile; (S.E.A.); (C.G.); (D.A.); (K.F.); (K.A.)
| | - Karen Flores
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1100000, Chile; (S.E.A.); (C.G.); (D.A.); (K.F.); (K.A.)
| | - Karem Arriaza
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1100000, Chile; (S.E.A.); (C.G.); (D.A.); (K.F.); (K.A.)
| |
Collapse
|
7
|
Kandy AT, Chand J, Baba MZ, Subramanian G. Is SIRT3 and Mitochondria a Reliable Target for Parkinson's Disease and Aging? A Narrative Review. Mol Neurobiol 2024:10.1007/s12035-024-04486-w. [PMID: 39287746 DOI: 10.1007/s12035-024-04486-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: 03/13/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Aging is a complicated degenerative process that has been thoroughly researched in a variety of taxa, including mammals, worms, yeast, and flies. One important controller of organismal lifetime is the conserved deacetylase protein known as silencing information regulator 2 (SIR2). It has been demonstrated that overexpressing SIR2 lengthens the life span in worms, flies, and yeast, demonstrating its function in enhancing longevity. SIRT3 is a member of the sirtuin protein family, identified as a major regulator of longevity and aging. Sirtuin 3 (SIRT3), a possible mitochondrial tumor suppressor, has been explicitly linked to the control of cellular reactive oxygen species (ROS) levels, the Warburg effect, and carcinogenesis. SIRT3 plays a significant part in neurodegenerative illnesses such as Parkinson's and Alzheimer's disease by decreasing the oxidative stress in mitochondria and reducing the ROS levels. Furthermore, SIRT3 has been linked to metabolic and cardiovascular disorders, indicating its wider role in the pathophysiology of disease and possible therapeutic applications.
Collapse
Affiliation(s)
- Amarjith Thiyyar Kandy
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India
| | - Jagdish Chand
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India
| | - Mohammad Zubair Baba
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India
| | - Gomathy Subramanian
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India.
| |
Collapse
|
8
|
Đorđević DB, Koračević GP, Đorđević AD, Lović DB. Hypertension and left ventricular hypertrophy. J Hypertens 2024; 42:1505-1515. [PMID: 38747417 DOI: 10.1097/hjh.0000000000003774] [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: 08/03/2024]
Abstract
In the initial stage, left ventricular hypertrophy (LVH) is adaptive, but in time, it transforms to maladaptive LVH which is specific for the development of various phenotypes that cause heart failure, initially with preserved, but later with reduced left ventricular ejection fraction. Pathophysiological mechanisms, which are characteristic for remodeling procedure, are numerous and extremely complex, and should be subjected to further research with the aim of making a comprehensive overview of hypertensive heart disease (HHD) and discovering new options for preventing and treating HHD. The contemporary methods, such as cardiac magnetic resonance (CMR) and computed tomography (CT) provide very accurate morphological and functional information on HHD. The objective of this review article is to summarize the available scientific information in terms of prevalence, pathophysiology, diagnostics, prevention, contemporary therapeutic options, as well as to present potential therapeutic solutions based on the research of pathological mechanisms which are at the core of HHD.
Collapse
Affiliation(s)
- Dragan B Đorđević
- Faculty of Medicine, University of Nis
- Institute for Treatment and Rehabilitation Niska Banja
| | - Goran P Koračević
- Faculty of Medicine, University of Nis
- Department for Cardiovascular Diseases, Clinical Center Nis, Nis, Serbia
| | | | - Dragan B Lović
- Clinic for Internal Diseases Intermedica, Singidunum University Nis, Jovana Ristica, Nis, Serbia
- Veterans Affair Medical Centre, Washington DC, USA
| |
Collapse
|
9
|
Fujii T, Ikari Y. Epidemiological and Prognostic Importance of New-Onset Cancer as a Net Adverse Clinical Outcome after ST-Elevation Myocardial Infarction. J Cardiovasc Dev Dis 2024; 11:256. [PMID: 39330314 PMCID: PMC11432219 DOI: 10.3390/jcdd11090256] [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: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
The study assessed the epidemiological frequency and prognostic impact of new-onset cancer as an additional net adverse clinical outcome in patients after ST-elevation myocardial infarction (STEMI), considering its potential clinical significance alongside classical endpoints. This study was designed as a single-center observational study, including 1285 consecutive patients who were diagnosed as STEMI patients as the subject, and the frequency and prognosis of new-onset cancer after STEMI onset were assessed. The incidence of all-cause death, nonfatal myocardial infarction (MI), stroke, and bleeding were analyzed as classical endpoints. Throughout an average of a 1241.4 days observation period, cancers were observed in 7.0% of patients (n = 90), showing development at a constant rate throughout this period (incidence rate, 0.06/1000 person-years). The average duration from STEMI onset to cancer diagnosis was 1371.4 days. Death, MI, or stroke were observed in 21.3%, 4.0%, 6.5%, and 12.8%, giving incidence rates of 0.18, 0.03, 0.06, and 0.11/1000 person-years, respectively. Long-term mortality was higher in patients with newly diagnosed cancer than in patients without cancer (36.7% vs. 20.1%, p < 0.01). Cancer after STEMI should be considered as an additional major adverse clinical event because of its high incidence, constant development, and high mortality in comparison to classical endpoints.
Collapse
Affiliation(s)
- Toshiharu Fujii
- Department of Cardiovascular Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | | |
Collapse
|
10
|
Smyła-Gruca W, Szczurek-Wasilewicz W, Skrzypek M, Romuk E, Karmański A, Jurkiewicz M, Gąsior M, Osadnik T, Banach M, Jóźwiak JJ, Szyguła-Jurkiewicz B. Ceruloplasmin and Lipofuscin Serum Concentrations Are Associated with Presence of Hypertrophic Cardiomyopathy. Biomedicines 2024; 12:1767. [PMID: 39200231 PMCID: PMC11352126 DOI: 10.3390/biomedicines12081767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/28/2024] [Accepted: 08/03/2024] [Indexed: 09/02/2024] Open
Abstract
Oxidative stress reflects an imbalance between the systemic manifestation of reactive oxygen species and cells' ability to neutralize them by antioxidant systems. The role of oxidative stress in hypertrophic cardiomyopathy (HCM) is not fully understood. The aim of the study was to examine selected parameters of oxidative stress in patients with HCM compared to the control group. We enrolled 85 consecutive HCM patients and 97 controls without HCM. The groups were matched for sex, the body mass index, and age. Oxidative stress markers included superoxide dismutase (SOD), ceruloplasmin (CER), and lipofuscin (LPS). The median age of the HCM patients was 53 (40-63) years, and 41.2% of them were male. HCM patients, compared to the control ones, had significantly increased levels of CER and LPS. The areas under the receiver operating characteristics curves (AUC) indicated a good discriminatory power of CER (AUC 0.924, sensitivity 84%, and specificity 88%), an acceptable discriminatory power of LPS (AUC 0.740, sensitivity 66%, and specificity 72%), and poor discriminatory power of SOD (AUC 0.556, sensitivity 34%, and specificity 94%) for HCM detection. CER with good predictive strength, as well as LPS with acceptable predictive power, allows for HCM detection. The utility of SOD for HCM detection is limited.
Collapse
Affiliation(s)
- Wiktoria Smyła-Gruca
- Student’s Scientific Society, 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (W.S.-G.); (M.J.)
| | | | - Michał Skrzypek
- Department of Biostatistics, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland;
| | - Ewa Romuk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Andrzej Karmański
- Department of Descriptive and Topographic Anatomy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Michał Jurkiewicz
- Student’s Scientific Society, 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (W.S.-G.); (M.J.)
| | - Mariusz Gąsior
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (M.G.); (B.S.-J.)
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
- Cardiology and Lipid Disorders Clinic, Independent Public Health Care Institution “REPTY” Upper Silesian Rehabilitation Centre, 42-600 Tarnowskie Góry, Poland
| | - Maciej Banach
- Polish Mothers Memorial Hospital Research Institute, 90-419 Łódź, Poland;
- Department of Hypertension, Medical University of Lodz, 90-419 Łódź, Poland
- Cardiovascular Research Centre, University of Zielona Gora, 65-046 Zielona Gora, Poland
| | - Jacek J. Jóźwiak
- Department of Family Medicine and Public Health, Faculty of Medicine, University of Opole, 45-040 Opole, Poland;
| | - Bożena Szyguła-Jurkiewicz
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (M.G.); (B.S.-J.)
| |
Collapse
|
11
|
Seth J, Sharma S, Leong CJ, Rabkin SW. Eicosapentaenoic Acid (EPA) and Docosahexaenoic Acid (DHA) Ameliorate Heart Failure through Reductions in Oxidative Stress: A Systematic Review and Meta-Analysis. Antioxidants (Basel) 2024; 13:955. [PMID: 39199201 PMCID: PMC11351866 DOI: 10.3390/antiox13080955] [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: 06/17/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/01/2024] Open
Abstract
The objectives of this study were to explore the role that eicosapentaenoic acid (EPA) and/or docosahexaenoic acid (DHA) plays in heart failure (HF), highlighting the potential connection to oxidative stress pathways. Following PRISMA guidelines, we conducted electronic searches of the literature in MEDLINE and EMBASE focusing on serum EPA and/or DHA and EPA and/or DHA supplementation in adult patients with heart failure or who had heart failure as an outcome of this study. We screened 254 studies, encompassing RCTs, observational studies, and cohort studies that examined HF outcomes in relation to either serum concentrations or dietary supplementation of EPA and/or DHA. The exclusion criteria were pediatric patients, non-HF studies, abstracts, editorials, case reports, and reviews. Eleven studies met our criteria. In meta-analyses, high serum concentrations of DHA were associated with a lower rate of heart failure with a hazard ratio of 0.74 (CI = 0.59-0.94). High serum concentrations of EPA also were associated with an overall reduction in major adverse cardiovascular events with a hazard ratio of 0.60 (CI = 0.46-0.77). EPA and DHA, or n3-PUFA administration, were associated with an increased LVEF with a mean difference of 1.55 (CI = 0.07-3.03)%. A potential explanation for these findings is the ability of EPA and DHA to inhibit pathways by which oxidative stress damages the heart or impairs cardiac systolic or diastolic function producing heart failure. Specifically, EPA may lower oxidative stress within the heart by reducing the concentration of reactive oxygen species (ROS) within cardiac tissue by (i) upregulating nuclear factor erythroid 2-related factor 2 (Nrf2), which increases the expression of antioxidant enzyme activity, including heme oxygenase-1, thioredoxin reductase 1, ferritin light chain, ferritin heavy chain, and manganese superoxide dismutase (SOD), (ii) increasing the expression of copper-zinc superoxide dismutase (MnSOD) and glutathione peroxidase, (iii) targeting Free Fatty Acid Receptor 4 (Ffar4), (iv) upregulating expression of heme-oxygenase-1, (v) lowering arachidonic acid levels, and (vi) inhibiting the RhoA/ROCK signaling pathway. DHA may lower oxidative stress within the heart by (i) reducing levels of mitochondrial-fission-related protein DRP-1(ser-63), (ii) promoting the incorporation of cardiolipin within the mitochondrial membrane, (iii) reducing myocardial fibrosis, which leads to diastolic heart failure, (iv) reducing the expression of genes such as Appa, Myh7, and Agtr1α, and (v) reducing inflammatory cytokines such as IL-6, TNF-α. In conclusion, EPA and/or DHA have the potential to improve heart failure, perhaps mediated by their ability to modulate oxidative stress.
Collapse
Affiliation(s)
- Jayant Seth
- Faculty of Medicine, University of British Columbia, 9th Floor 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada; (J.S.); (S.S.); (C.J.L.)
| | - Sohat Sharma
- Faculty of Medicine, University of British Columbia, 9th Floor 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada; (J.S.); (S.S.); (C.J.L.)
| | - Cameron J. Leong
- Faculty of Medicine, University of British Columbia, 9th Floor 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada; (J.S.); (S.S.); (C.J.L.)
| | - Simon W. Rabkin
- Faculty of Medicine, University of British Columbia, 9th Floor 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada; (J.S.); (S.S.); (C.J.L.)
- Department of Medicine, Division of Cardiology, University of British Columbia, 9th Floor 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
12
|
Jing B, Chen ZN, Si WM, Zhao JJ, Zhao GP, Zhang D. (+)-Catechin attenuates CCI-induced neuropathic pain in male rats by promoting the Nrf2 antioxidant pathway to inhibit ROS/TLR4/NF-κB-mediated activation of the NLRP3 inflammasome. J Neurosci Res 2024; 102:e25372. [PMID: 39086264 DOI: 10.1002/jnr.25372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 07/06/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024]
Abstract
The objective of this study was to investigate the potential mechanisms by which (+)-catechin alleviates neuropathic pain. Thirty-two male Sprague-Dawley rats were divided into four groups: the sham group, the chronic constriction injury (CCI)group, the CCI+ ibuprofen group, and the CCI+ (+)-catechin group. CCI surgery induces thermal hyperalgesia in rats and (+)-catechin ameliorated CCI-induced thermal hyperalgesia and repaired damaged sciatic nerve in rats. CCI decreased SOD levels in male rat spinal cord dorsal horn and promoted MDA production, induced oxidative stress by increasing NOX4 levels and decreasing antioxidant enzyme HO-1 levels, and also increased protein levels of TLR4, p-NF-κB, NLRP3 inflammasome components, and IL-1β. In contrast, (+)-catechin reversed the above results. In i vitro experiments, (+)-catechin reduced the generation of reactive oxygen species (ROS) in GMI-R1 cells after LPS stimulation and attenuated the co-expression of IBA-1 and NLRP3. It also showed significant inhibition of the NF-κB and NLRP3 inflammatory pathways and activation of the Nrf2-mediated antioxidant system. Overall, these findings suggest that (+)-catechin inhibits the activation of the NLRP3 inflammasome through the triggering of the Nrf2-induced antioxidant system, the inhibition of the TLR4/NF-κB pathway, and the production of ROS to alleviate CCI-induced neuropathic pain in male rats.
Collapse
Affiliation(s)
- Bei Jing
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhen-Ni Chen
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wai-Mei Si
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jia-Ji Zhao
- Chemistry & Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guo-Ping Zhao
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Di Zhang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
13
|
Samavati I, Ranjbar A, Haddadi R. Cardioprotective effect of vitamin D3 on cisplatin-induced cardiotoxicity in male mice: role of oxidative stress. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4761-4769. [PMID: 38150016 DOI: 10.1007/s00210-023-02848-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/09/2023] [Indexed: 12/28/2023]
Abstract
Cisplatin (CP) is a chemotherapy drug used in a broad spectrum of cancer. The current study investigated the protective effect of vitamin D3 (vit-D3) on CP-induced cardiotoxicity. Forty-two male Balb-c mice (20-25 g) were divided into seven groups (GP), 6 per/group were included: GP1 was considered the control group, GP2 received a single dose of I.V. injection of cisplatin (10 mg/kg). Seven days before cisplatin injection on GP3 and GP4 as pre-treatment, vit-D3 was injected I.P. with the doses of 500 IU/kg and 1000 IU/kg, respectively. GP5 and GP6 were considered the treatment groups, were injected cisplatin (10 mg/kg, I.V), and 15 days later, received vit-D3 (500 IU/kg and 1000 IU/kg, I.P) for 7 days. GP7 was the positive control group, which received vit-D3 at a dose of 500 IU/kg (I.P.) for 7 days. Tissues samples and blood serum were collected for biochemical and histopathological investigations. CP injection significantly increased (p < 0.001) LDH, Troponin I, CK-MB, malondialdehyde (MDA), and nitric oxide (NO) levels, but total antioxidant capacity (TAC) levels were significantly reduced. Histological findings showed cardiac muscle rupture, myocardial fiber necrosis, edema, and pyknotic nuclei, indicating cardiac damage. In both pre-treatment and treatment protocol, vit-D3 could improve the histological and biochemical parameters and prevented from the CP toxicity. Vit-D3 significantly could prevent the CP cardiotoxicity in pre-treatment groups, and partially improve the damage of chemotherapy in treatment group. However, further research is necessary to establish the potential of vit-D3 in preventing or ameliorating cisplatin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Iman Samavati
- Department of Pharmacology and Toxicology, School of Pharmacy, Herbal Medicine and Natural Product Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Akram Ranjbar
- Department of Pharmacology and Toxicology, School of Pharmacy, Herbal Medicine and Natural Product Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Rasool Haddadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Herbal Medicine and Natural Product Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran.
| |
Collapse
|
14
|
Julovi SM, Trinh K, Robertson H, Xu C, Minhas N, Viswanathan S, Patrick E, Horowitz JD, Meijles DN, Rogers NM. Thrombospondin-1 Drives Cardiac Remodeling in Chronic Kidney Disease. JACC Basic Transl Sci 2024; 9:607-627. [PMID: 38984053 PMCID: PMC11228122 DOI: 10.1016/j.jacbts.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 07/11/2024]
Abstract
Patients with chronic kidney disease (CKD) face a high risk of cardiovascular disease. Previous studies reported that endogenous thrombospondin 1 (TSP1) involves right ventricular remodeling and dysfunction. Here we show that a murine model of CKD increased myocardial TSP1 expression and produced left ventricular hypertrophy, fibrosis, and dysfunction. TSP1 knockout mice were protected from these features. In vitro, indoxyl sulfate is driving deleterious changes in cardiomyocyte through the TSP1. In patients with CKD, TSP1 and aryl hydrocarbon receptor were both differentially expressed in the myocardium. Our findings summon large clinical studies to confirm the translational role of TSP1 in patients with CKD.
Collapse
Affiliation(s)
- Sohel M Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Harry Robertson
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
| | - Cuicui Xu
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Nikita Minhas
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Seethalakshmi Viswanathan
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Ellis Patrick
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
- School of Mathematics, University of Sydney, New South Wales, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong Special Administrative Region, China
| | - John D Horowitz
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
- Cardiovascular Pathophysiology and Therapeutics Research Group, Basil Hetzel Institute, Woodville, South Australia, Australia
- Department of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Natasha M Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Renal and Transplantation Unit, Westmead Hospital, New South Wales, Australia
| |
Collapse
|
15
|
Leite LB, Soares LL, Portes AMO, Soares TI, da Silva BAF, Dias TR, Costa SFF, Guimarães-Ervilha LO, Assis MQ, Lavorato VN, da Silva AN, Machado-Neves M, Reis ECC, Natali AJ. Combined physical training protects the left ventricle from structural and functional damages in experimental pulmonary arterial hypertension. Clin Hypertens 2024; 30:12. [PMID: 38689333 PMCID: PMC11061945 DOI: 10.1186/s40885-024-00270-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Under the adverse remodeling of the right ventricle and interventricular septum in pulmonary arterial hypertension (PAH) the left ventricle (LV) dynamics is impaired. Despite the benefits of combined aerobic and resistance physical trainings to individuals with PAH, its impact on the LV is not fully understood. OBJECTIVE To test whether moderate-intensity combined physical training performed during the development of PAH induced by MCT in rats is beneficial to the LV's structure and function. METHODS Male Wistar rats were divided into two groups: Sedentary Hypertensive Survival (SHS, n = 7); and Exercise Hypertensive Survival (EHS, n = 7) to test survival. To investigate the effects of combined physical training, another group of rats were divided into three groups: Sedentary Control (SC, n = 7); Sedentary Hypertensive (SH, n = 7); and Exercise Hypertensive (EH, n = 7). PAH was induced through an intraperitoneal injection of MCT (60 mg/kg). Echocardiographic evaluations were conducted on the 22nd day after MCT administration. Animals in the EHS and EH groups participated in a combined physical training program, alternating aerobic (treadmill running: 50 min, 60% maximum running speed) and resistance (ladder climbing: 15 climbs with 1 min interval, 60% maximum carrying load) exercises, one session/day, 5 days/week for approximately 4 weeks. RESULTS The physical training increased survival and tolerance to aerobic (i.e., maximum running speed) and resistance (i.e., maximum carrying load) exertions and prevented reductions in ejection fraction and fractional shortening. In addition, the physical training mitigated oxidative stress (i.e., CAT, SOD and MDA) and inhibited adverse LV remodeling (i.e., Collagen, extracellular matrix, and cell dimensions). Moreover, the physical training preserved the amplitude and velocity of contraction and hindered the reductions in the amplitude and velocity of the intracellular Ca2+ transient in LV single myocytes. CONCLUSION Moderate-intensity combined physical training performed during the development of MCT-induced PAH in rats protects their LV from damages to its structure and function and hence increases their tolerance to physical exertion and prolongs their survival.
Collapse
Affiliation(s)
- Luciano Bernardes Leite
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil.
| | - Leôncio Lopes Soares
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | - Thayana Inácia Soares
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | - Taís Rodrigues Dias
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | - Luiz Otávio Guimarães-Ervilha
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Mirian Quintão Assis
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Victor Neiva Lavorato
- Department of Physical Education, Governador Ozanam Coelho University Center, Ubá, Minas Gerais, Brazil
| | | | - Mariana Machado-Neves
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Antônio José Natali
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| |
Collapse
|
16
|
Zhang L, Xie F, Zhang F, Lu B. The potential roles of exosomes in pathological cardiomyocyte hypertrophy mechanisms and therapy: A review. Medicine (Baltimore) 2024; 103:e37994. [PMID: 38669371 PMCID: PMC11049793 DOI: 10.1097/md.0000000000037994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Pathological cardiac hypertrophy, characterized by the enlargement of cardiac muscle cells, leads to serious cardiac conditions and stands as a major global health issue. Exosomes, comprising small lipid bilayer vesicles, are produced by various cell types and found in numerous bodily fluids. They play a pivotal role in intercellular communication by transferring bioactive cargos to recipient cells or activating signaling pathways in target cells. Exosomes from cardiomyocytes, endothelial cells, fibroblasts, and stem cells are key in regulating processes like cardiac hypertrophy, cardiomyocyte survival, apoptosis, fibrosis, and angiogenesis within the context of cardiovascular diseases. This review delves into exosomes' roles in pathological cardiac hypertrophy, first elucidating their impact on cell communication and signaling pathways. It then advances to discuss how exosomes affect key hypertrophic processes, including metabolism, fibrosis, oxidative stress, and angiogenesis. The review culminates by evaluating the potential of exosomes as biomarkers and their significance in targeted therapeutic strategies, thus emphasizing their critical role in the pathophysiology and management of cardiac hypertrophy.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Xie
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fengmei Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Beiyao Lu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Sørhus E, Bjelland R, Durif C, Johnsen E, Donald CE, Meier S, Nordtug T, Vikebø FB, Perrichon P. Oil droplet fouling on lesser sandeel (Ammodytes marinus) eggshells does not enhance the crude oil induced developmental toxicity. JOURNAL OF HAZARDOUS MATERIALS 2024; 468:133814. [PMID: 38412802 DOI: 10.1016/j.jhazmat.2024.133814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/29/2024]
Abstract
The oil industry's expansion and increased operational activity at older installations, along with their demolition, contribute to rising cumulative pollution and a heightened risk of accidental oil spills. The lesser sandeel (Ammodytes marinus) is a keystone prey species in the North Sea and coastal systems. Their eggs adhere to the seabed substrate making them particularly vulnerable to oil exposure during embryonic development. We evaluated the sensitivity of sandeel embryos to crude oil in a laboratory by exposing them to dispersed oil at concentrations of 0, 15, 50, and 150 µg/L oil between 2 and 16 days post-fertilization. We assessed water and tissue concentrations of THC and tPAH, cyp1a expression, lipid distribution in the eyes, head and trunk, and morphological and functional deformities. Oil droplets accumulated on the eggshell in all oil treatment groups, to which the embryo responded by a dose-dependent rise in cyp1a expression. The oil exposure led to only minor sublethal deformities in the upper jaw and otic vesicle. The findings suggest that lesser sandeel embryos are resilient to crude oil exposure. The lowest observed effect level documented in this study was 36 µg THC/L and 3 µg tPAH/L. The inclusion of these species-specific data in risk assessment models will enhance the precision of risk evaluations for the North Atlantic ecosystems.
Collapse
Affiliation(s)
- Elin Sørhus
- Institute of Marine Research, Bergen, Norway.
| | - Reidun Bjelland
- Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Caroline Durif
- Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | | | | | | | | | | | | |
Collapse
|
18
|
Yu S, Xiong L, Wei D, Zhu H, Cai X, Shao L, Hong L, Zhan Y. Prediction of the left ventricular mass index in hypertensive patients using the product of red cell distribution width and mean corpuscular volume. Medicine (Baltimore) 2024; 103:e37685. [PMID: 38579056 PMCID: PMC10994413 DOI: 10.1097/md.0000000000037685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/01/2024] [Indexed: 04/07/2024] Open
Abstract
The product of red cell distribution width (RDW) and mean corpuscular volume (MCV) has been identified as an indicator of target organ damage in cases of hypertension. However, the role of the RDW-MCV product in assessing carotid alteration, renal damage, and left ventricular hypertrophy in patients with hypertension has not been elucidated. In this cross-sectional study, a total of 1115 participants with hypertension were included. The RDW and MCV at admission were measured using an automated hematology analyzer. Organ damage was determined by the left ventricular mass index (LVMI), carotid intima-media thickness, and estimated glomerular filtration rate. The prevalence rates of carotid alteration and left ventricular hypertrophy were 57.0% and 18.0%, respectively. A higher RDW-MCV product and RDW were observed in hypertensive patients who developed carotid alteration. After adjusting for potential confounding factors, the correlations of the RDW-MCV product (P = .285) and RDW (P = .346) with carotid alteration were not significant. Moreover, the analysis of variance showed no significant correlation between RDW and LVMI (P = .186). However, the RDW-MCV product was higher in individuals with a high LVMI compared to those with a normal LVMI. Multivariable linear regression analysis revealed that the RDW-MCV product was independently associated with the LVMI (β = 2.519, 95% CI: 0.921-4.116; P = .002), but not the estimated glomerular filtration rate (β = -0.260, 95% CI: -2.031-1.511; P = .773). An elevated RDW-MCV product may be a predictor for left ventricular hypertrophy in patients with hypertension.
Collapse
Affiliation(s)
- Songping Yu
- Department of Cardiology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Lingbing Xiong
- The Second Department of Cardiology, The Third Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Dan Wei
- Department of Cardiology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Hongmin Zhu
- Department of Cardiology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Xinyong Cai
- Department of Cardiology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Liang Shao
- Department of Cardiology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Lang Hong
- Department of Cardiology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yuliang Zhan
- Department of Cardiology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Zhang Y, Huang K, Duan J, Zhao R, Yang L. Gut microbiota connects the brain and the heart: potential mechanisms and clinical implications. Psychopharmacology (Berl) 2024; 241:637-651. [PMID: 38407637 DOI: 10.1007/s00213-024-06552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/04/2024] [Indexed: 02/27/2024]
Abstract
Nowadays, high morbidity and mortality of cardiovascular diseases (CVDs) and high comorbidity rate of neuropsychiatric disorders contribute to global burden of health and economics. Consequently, a discipline concerning abnormal connections between the brain and the heart and the resulting disease states, known as psychocardiology, has garnered interest among researchers. However, identifying a common pathway that physicians can modulate remains a challenge. Gut microbiota, a constituent part of the human intestinal ecosystem, is likely involved in mutual mechanism CVDs and neuropsychiatric disorder share, which could be a potential target of interventions in psychocardiology. This review aimed to discuss complex interactions from the perspectives of microbial and intestinal dysfunction, behavioral factors, and pathophysiological changes and to present possible approaches to regulating gut microbiota, both of which are future directions in psychocardiology.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Kai Huang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Jiahao Duan
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Rong Zhao
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| |
Collapse
|
20
|
Teng Y, Li Y, Wang L, Wang B, Su S, Chen J, Lu Z, Zhu H, Zhao M. Effectiveness and pharmacological mechanisms of Chinese herbal medicine for coronary heart disease complicated with heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117605. [PMID: 38128892 DOI: 10.1016/j.jep.2023.117605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine (CHM) is widely used for treating coronary heart disease complicated with heart failure (CHD-HF). However, the exact mechanisms involved are still not fully understood. AIM OF THE STUDY To assess the clinical effectiveness and potential pharmacological mechanisms of CHM for treating CHD-HF. METHODS Eight databases were retrieved for Randomized Controlled Trials of CHM for CHD-HF published from their inception to March 2023. Quality assessment of include studies was performed by the Cochrane risk-of-bias. Meta-analysis was used to assess the effectiveness of CHM for CHD-HF, and then core drugs and active ingredients were selected by data mining and network pharmacology. Finally, cluster and enrichment analysis were adopted to explore the potential targets and signaling pathways. RESULTS A total of 52 studies enrolling 5216 patients were included. Meta-analysis revealed that CHM treatment groups significantly improved left ventricular ejection fraction (LVEF), 6-min walk test (6-MWT), left ventricular end-diastolic dimension (LVEDD) and left ventricular end systolic diameter (LVESD) than control groups: [LVEF: SMD = 0.7, 95%CI (0.54, 0.87), p < 0.00001, I2 = 80%; 6-MWT: SMD = 0.72, 95%CI (0.58, 0.86), p < 0.0001, I2 = 67%; LVEDD: SMD = -0.79, 95%CI (-0.89, -0.69), p < 0.0001, I2 = 49%; LVESD: SMD = -0.6 (-0.74, -0.46), p < 0.0001, I2 = 0%]. The results of various biological information analysis showed the internal relationship between prescriptions, core drugs, active ingredients and therapeutic targets. Twelve core herbs with the most commonly use and high correlation were selected from 110 CHMs of 52 prescriptions for CHD-HF treatment, and further 65 effective components were screened out according to the most strength value, which were divided into 12 compounds such as terpenoids, flavonoids, steroids and alkaloids and etc. At the same time, 67 therapeutic targets of active ingredients in CHD-HF were filtrated. On these bases, cluster and enrichment analysis of the components and targets were used to explore relevant pharmacological mechanisms, mainly including anti-myocardial cell damage, anti-inflammation, anti-apoptosis, anti-fibrosis, regulation of oxidative stress, anticoagulation and angiogenesis, and improvement of glucose and fatty acid metabolism. CONCLUSION CHM are effective in treating CHD-HF compared with conventional treatment. Some of the included studies have high risks in the implementation of blinding, so more high-quality studies are needed. The active ingredients of CHM could protect the myocardium and improve pathological environment of CHD-HF in various ways. And CHM has the advantage of multi-component and multi-target treatment for complex diseases.
Collapse
Affiliation(s)
- Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jiaxin Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Haiyan Zhu
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Institute of Cardiovascular Diseases, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
21
|
Edwards CV, Ferri GM, Villegas-Galaviz J, Ghosh S, Bawa PS, Wang F, Klimtchuk E, Ajayi TB, Morgan GJ, Prokaeva T, Staron A, Ruberg FL, Sanchorawala V, Giadone RM, Murphy GJ. Abnormal global longitudinal strain and reduced serum inflammatory markers in cardiac AL amyloidosis patients without significant amyloid fibril deposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.584987. [PMID: 38558967 PMCID: PMC10980073 DOI: 10.1101/2024.03.14.584987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background Cardiac dysfunction in AL amyloidosis is thought to be partly related to the direct impact of AL LCs on cardiomyocyte function, with the degree of dysfunction at diagnosis as a major determinant of clinical outcomes. Nonetheless, mechanisms underlying LC-induced myocardial toxicity are not well understood. Methods We identified gene expression changes correlating with human cardiac cells exposed to a cardiomyopathy-associated κAL LC. We then sought to confirm these findings in a clinical dataset by focusing on clinical parameters associated with the pathways dysregulated at the gene expression level. Results Upon exposure to a cardiomyopathy-associated κAL LC, cardiac cells exhibited gene expression changes related to myocardial contractile function and inflammation, leading us to hypothesize that there could be clinically detectable changes in GLS on echocardiogram and serum inflammatory markers in patients. Thus, we identified 29 patients with normal IVSd but abnormal cardiac biomarkers suggestive of LC-induced cardiac dysfunction. These patients display early cardiac biomarker staging, abnormal GLS, and significantly reduced serum inflammatory markers compared to patients with clinically evident amyloid fibril deposition. Conclusion Collectively, our findings highlight early molecular and functional signatures of cardiac AL amyloidosis, with potential impact for developing improved patient biomarkers and novel therapeutics.
Collapse
|
22
|
Wong CN, Gui XY, Rabkin SW. Myeloperoxidase, carnitine, and derivatives of reactive oxidative metabolites in heart failure with preserved versus reduced ejection fraction: A meta-analysis. Int J Cardiol 2024; 399:131657. [PMID: 38101703 DOI: 10.1016/j.ijcard.2023.131657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/03/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND Understanding the pathophysiology of heart failure (HF) with preserved ejection fraction (HFpEF) continues to be challenging. Several inflammatory and metabolic biomarkers have recently been suggested to be involved in HFpEF. OBJECTIVES The purpose of this review was to synthesize the evidence on non-traditional biomarkers from metabolomic studies that may distinguish HFpEF from heart failure with reduced ejection fraction (HFrEF) and controls without HF. METHODS A systematic search was conducted using Medline and PubMed with search terms such as "HFpEF" and "metabolomics", and a meta-analysis was conducted. RESULTS Myeloperoxidase (MPO) levels were significantly (p < 0.001) higher in HFpEF than controls without HF, but comparable (p = 0.838) between HFpEF and HFrEF. Carnitine levels were significantly (p < 0.0001) higher in HFrEF than HFpEF, but comparable (p = 0.443) between HFpEF and controls without HF. Derivatives of reactive oxidative metabolites (DROMs) were not significantly (p = 0.575) higher in HFpEF than controls without HF. CONCLUSION These data suggest that MPO is operative in HFpEF and HFrEF and may be a biomarker for HF. Furthermore, circulating carnitine levels may distinguish HFrEF from HFpEF.
Collapse
Affiliation(s)
- Chenille N Wong
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Xi Yao Gui
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Simon W Rabkin
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Division of Cardiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
23
|
Shi X, Dang X, Huang Z, Lu Y, Tong H, Liang F, Zhuang F, Li Y, Cai Z, Huo H, Jiang Z, Pan C, Wang X, Gu C, He B. SUMOylation of TEAD1 Modulates the Mechanism of Pathological Cardiac Hypertrophy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305677. [PMID: 38225750 PMCID: PMC10966521 DOI: 10.1002/advs.202305677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/14/2023] [Indexed: 01/17/2024]
Abstract
Pathological cardiac hypertrophy is the leading cause of heart failure and has an extremely complicated pathogenesis. TEA domain transcription factor 1 (TEAD1) is recognized as an important transcription factor that plays a key regulatory role in cardiovascular disease. This study aimed to explore the role of TEAD1 in cardiac hypertrophy and to clarify the regulatory role of small ubiquitin-like modifier (SUMO)-mediated modifications. First, the expression level of TEAD1 in patients with heart failure, mice, and cardiomyocytes is investigated. It is discovered that TEAD1 is modified by SUMO1 during cardiac hypertrophy and that the process of deSUMOylation is regulated by SUMO-specific protease 1 (SENP1). Lysine 173 is an essential site for TEAD1 SUMOylation, which affects the protein stability, nuclear localization, and DNA-binding ability of TEAD1 and enhances the interaction between TEAD1 and its transcriptional co-activator yes-associated protein 1 in the Hippo pathway. Finally, adeno-associated virus serotype 9 is used to construct TEAD1 wild-type and KR mutant mice and demonstrated that the deSUMOylation of TEAD1 markedly exacerbated cardiomyocyte enlargement in vitro and in a mouse model of cardiac hypertrophy. The results provide novel evidence that the SUMOylation of TEAD1 is a promising therapeutic strategy for hypertrophy-related heart failure.
Collapse
Affiliation(s)
- Xin Shi
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Xuening Dang
- Department of Cardiovascular SurgeryShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Zhenyu Huang
- Department of Central LaboratoryShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Yanqiao Lu
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Huan Tong
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Feng Liang
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Fei Zhuang
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Yi Li
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Zhaohua Cai
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Huanhuan Huo
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Zhaolei Jiang
- Department of Cardiothoracic SurgeryXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200030China
| | - Changqing Pan
- General Surgery DepartmentShanghai Chest HospitalSchool of Medicine Shanghai Jiao Tong UniversityShanghai200030China
| | - Xia Wang
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Chang Gu
- Department of Cardiothoracic SurgeryXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200030China
- Department of Thoracic SurgeryShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Ben He
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| |
Collapse
|
24
|
Cinato M, Andersson L, Miljanovic A, Laudette M, Kunduzova O, Borén J, Levin MC. Role of Perilipins in Oxidative Stress-Implications for Cardiovascular Disease. Antioxidants (Basel) 2024; 13:209. [PMID: 38397807 PMCID: PMC10886189 DOI: 10.3390/antiox13020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Oxidative stress is the imbalance between the production of reactive oxygen species (ROS) and antioxidants in a cell. In the heart, oxidative stress may deteriorate calcium handling, cause arrhythmia, and enhance maladaptive cardiac remodeling by the induction of hypertrophic and apoptotic signaling pathways. Consequently, dysregulated ROS production and oxidative stress have been implicated in numerous cardiac diseases, including heart failure, cardiac ischemia-reperfusion injury, cardiac hypertrophy, and diabetic cardiomyopathy. Lipid droplets (LDs) are conserved intracellular organelles that enable the safe and stable storage of neutral lipids within the cytosol. LDs are coated with proteins, perilipins (Plins) being one of the most abundant. In this review, we will discuss the interplay between oxidative stress and Plins. Indeed, LDs and Plins are increasingly being recognized for playing a critical role beyond energy metabolism and lipid handling. Numerous reports suggest that an essential purpose of LD biogenesis is to alleviate cellular stress, such as oxidative stress. Given the yet unmet suitability of ROS as targets for the intervention of cardiovascular disease, the endogenous antioxidant capacity of Plins may be beneficial.
Collapse
Affiliation(s)
- Mathieu Cinato
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Linda Andersson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Azra Miljanovic
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Marion Laudette
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University—Paul Sabatier, 31432 Toulouse, France;
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Malin C. Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| |
Collapse
|
25
|
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: 0] [Impact Index Per Article: 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
|
26
|
Zampieri M, Schoonvelde SAC, Vinci M, Meattini I, Visani L, Fornaro A, Coppini R, Romei A, Marchi A, Morelli I, van Slegtenhorst MA, Palinkas ED, Livi L, Michels M, Olivotto I. Cancer Treatment-Related Complications in Patients With Hypertrophic Cardiomyopathy. Mayo Clin Proc 2024; 99:218-228. [PMID: 38180395 DOI: 10.1016/j.mayocp.2023.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 07/25/2023] [Accepted: 10/05/2023] [Indexed: 01/06/2024]
Abstract
OBJECTIVE To describe the potential clinical cardiotoxicity of oncological treatments in a cohort of consecutive patients with hypertrophic cardiomyopathy (HCM), systematically followed-up at two national referral centers for HCM. Cardiotoxicity relates to the direct effects of cancer-related treatment on heart function, commonly presenting as left ventricular contractile dysfunction. However, limited data are available regarding cardiotoxic effects on HCM as most studies have not specifically analyzed the effects of oncological treatment in HCM populations. This gap in knowledge may lead to unjustified restriction of HCM patients from receiving curative cancer treatments. METHODS We retrospectively analyzed clinical and instrumental data of all consecutive HCM patients who underwent oncological treatment between January 2000 and December 2020 collected in a centralized database. RESULTS Of 3256 HCM patients, 121 (3.7%) had cancer; 110 (90.9%) underwent oncological surgery, 45 (37.2%) received chemotherapy, and 22 (18.2%) received chest radiation therapy (cRT). After a median follow-up of 5.2 years (Q1-Q3: 2-13 years) from oncological diagnosis, 32 patients died. The cumulative survival at 5 years was 79.9%. The cause of death was mainly attributed to the oncological condition, whereas four patients died of sudden cardiac death without receiving previous chemotherapy or cRT. No patient interrupted or reduced the dose of oncological treatment due to cardiac dysfunction. No sustained ventricular tachyarrhythmia was induced by chemotherapy or radiation therapy. CONCLUSION Cancer treatment was well tolerated in HCM patients. In our consecutive series, none died of cardiovascular complications induced by chemotherapy or cRT and they did not require interruption or substantial treatment tapering due to cardiovascular toxic effects. Although a multidisciplinary evaluation is necessary and regimens must be tailored individually, the diagnosis of HCM per se should not be considered a contraindication to receive optimal curative cancer treatment.
Collapse
Affiliation(s)
- Mattia Zampieri
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy; Pediatric Cardiology, Meyer Children's University Hospital IRCCS, Florence, Italy.
| | - Stephan A C Schoonvelde
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, Rotterdam, Netherlands
| | - Michele Vinci
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Icro Meattini
- Radiation Oncology Unit - Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Luca Visani
- Radiation Oncology Unit - Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | | | - Raffaele Coppini
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Andrea Romei
- Radiation Oncology Unit - Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Alberto Marchi
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Ilaria Morelli
- Radiation Oncology Unit - Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Marjon A van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Eszter Dalma Palinkas
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy; Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Lorenzo Livi
- Radiation Oncology Unit - Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Michelle Michels
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, Rotterdam, Netherlands
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy; Pediatric Cardiology, Meyer Children's University Hospital IRCCS, Florence, Italy; Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| |
Collapse
|
27
|
Marta CI, Craina M, Nitu R, Maghiari AL, Abu-Awwad SA, Boscu L, Diaconu M, Dumitru C, Dahma G, Yasar II, Babes K. A Comparative Analysis of NT-proBNP Levels in Pregnant Women and the Impact of SARS-CoV-2 Infection: Influence on Birth Outcome. Diseases 2023; 12:10. [PMID: 38248361 PMCID: PMC10814387 DOI: 10.3390/diseases12010010] [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: 11/29/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND The cardiac biomarker NT-proBNP is released by the ventricles in response to increased cardiac wall tension, showing cardiac activity in heart failure. The primary objective of this comparative study was to analyze the variations of NT-proBNP levels among pregnant patients and to determine the potential influence of SARS-CoV-2 infection on these values. Secondly, the study focused on NT-proBNP levels and their influence on the type of birth. METHODS Blood samples were taken from 160 pregnant mothers in order to determine, through the solid-phase enzyme-linked immunosorbent assay (ELISA) method, the NT-proBNP concentrations from the plasma. The cohort was separated into two distinct groups based on SARS-CoV-2 diagnostic results: negative to the infection, and positive to the infection. RESULTS The SARS-CoV-2-positive group of patients presented with higher levels of NT-proBNP and had higher rates of cesarean sections. (4) Conclusions: Our research highlights the crucial relationship between elevated NT-proBNP values and the mode of giving birth, natural delivery or cesarean section, and also the influence of SARS-CoV-2 viral infection and this biomarker.
Collapse
Affiliation(s)
- Carmen-Ioana Marta
- Doctoral School, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410087 Oradea, Romania;
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania; (M.C.); (M.D.); (C.D.); (G.D.)
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Marius Craina
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania; (M.C.); (M.D.); (C.D.); (G.D.)
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Razvan Nitu
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania; (M.C.); (M.D.); (C.D.); (G.D.)
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Anca Laura Maghiari
- Department I—Discipline of Anatomy and Embryology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Simona-Alina Abu-Awwad
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (S.-A.A.-A.); (L.B.); (I.-I.Y.)
| | - Lioara Boscu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (S.-A.A.-A.); (L.B.); (I.-I.Y.)
| | - Mircea Diaconu
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania; (M.C.); (M.D.); (C.D.); (G.D.)
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Catalin Dumitru
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania; (M.C.); (M.D.); (C.D.); (G.D.)
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - George Dahma
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania; (M.C.); (M.D.); (C.D.); (G.D.)
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (S.-A.A.-A.); (L.B.); (I.-I.Y.)
| | - Ionela-Iasmina Yasar
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (S.-A.A.-A.); (L.B.); (I.-I.Y.)
- Department IX: Surgery I, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Katalin Babes
- Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410087 Oradea, Romania;
- Clinical County Emergency Hospital of Oradea, 410167 Oradea, Romania
| |
Collapse
|
28
|
Karna S, Kang KW. An Overview of the Mechanism behind Excessive Volume of Pericardial Fat in Heart Failure. J Obes Metab Syndr 2023; 32:322-329. [PMID: 38036419 PMCID: PMC10786210 DOI: 10.7570/jomes23042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Heart failure (HF) is a clinical syndrome characterized by myocardial dysfunction leading to inefficient blood filling or ejection. Regardless of the etiology, various mechanisms, including adipokine hypersecretion, proinflammatory cytokines, stem cell proliferation, oxidative stress, hyperglycemic toxicity, and autonomic nervous system dysregulation in the pericardial fat (PCF), contribute to the development of HF. PCF has been directly associated with cardiovascular disease, and an increased PCF volume is associated with HF. The PCF acts as neuroendocrine tissue that is closely linked to myocardial function and acts as an energy reservoir. This review aims to summarize each mechanism associated with PCF in HF.
Collapse
Affiliation(s)
- Sandeep Karna
- Division of Cardiology, Cardiovascular Arrhythmia Center, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Ki-Woon Kang
- Division of Cardiology, Cardiovascular Arrhythmia Center, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
29
|
Qin C, Xu D, Han H, Fang J, Wang H, Liu Y, Wang H, Zhou X, Li D, Ying Y, Hu N, Xu L. Dynamic and Label-Free Sensing of Cardiomyocyte Responses to Nanosized Vesicles for Cardiac Oxidative Stress Injury Therapy. NANO LETTERS 2023; 23:11850-11859. [PMID: 38051785 DOI: 10.1021/acs.nanolett.3c03892] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Cardiac oxidative stress is a significant phenotype of myocardial infarction disease, a leading cause of global health threat. There is an urgent need to develop innovative therapies. Nanosized extracellular vesicle (nEV)-based therapy shows promise, yet real-time monitoring of cardiomyocyte responses to nEVs remains a challenge. In this study, a dynamic and label-free cardiomyocyte biosensing system using microelectrode arrays (MEAs) was constructed. Cardiomyocytes were cultured on MEA devices for electrophysiological signal detection and treated with nEVs from E. coli, gardenia, HEK293 cells, and mesenchymal stem cells (MSC), respectively. E. coli-nEVs and gardenia-nEVs induced severe paroxysmal fibrillation, revealing distinct biochemical communication compared to MSC-nEVs. Principal component analysis identified variations and correlations between nEV types. MSC-nEVs enhanced recovery without inducing arrhythmias in a H2O2-induced oxidative stress injury model. This study establishes a fundamental platform for assessing biochemical communication between nEVs and cardiomyocytes, offering new avenues for understanding nEVs' functions in the cardiovascular system.
Collapse
Affiliation(s)
- Chunlian Qin
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| | - Dongxin Xu
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Haote Han
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| | - Jiaru Fang
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Hao Wang
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Yingjia Liu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| | - Haobo Wang
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| | - Xin Zhou
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Danyang Li
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yibin Ying
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| | - Ning Hu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, Zhejiang University, Hangzhou 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Lizhou Xu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| |
Collapse
|
30
|
Cai L, Pessoa MT, Gao Y, Strause S, Banerjee M, Tian J, Xie Z, Pierre SV. The Na/K-ATPase α1/Src Signaling Axis Regulates Mitochondrial Metabolic Function and Redox Signaling in Human iPSC-Derived Cardiomyocytes. Biomedicines 2023; 11:3207. [PMID: 38137428 PMCID: PMC10740578 DOI: 10.3390/biomedicines11123207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Na/K-ATPase (NKA)-mediated regulation of Src kinase, which involves defined amino acid sequences of the NKA α1 polypeptide, has emerged as a novel regulatory mechanism of mitochondrial function in metazoans. Mitochondrial metabolism ensures adequate myocardial performance and adaptation to physiological demand. It is also a critical cellular determinant of cardiac repair and remodeling. To assess the impact of the proposed NKA/Src regulatory axis on cardiac mitochondrial metabolic function, we used a gene targeting approach in human cardiac myocytes. Human induced pluripotent stem cells (hiPSC) expressing an Src-signaling null mutant (A420P) form of the NKA α1 polypeptide were generated using CRISPR/Cas9-mediated genome editing. Total cellular Na/K-ATPase activity remained unchanged in A420P compared to the wild type (WT) hiPSC, but baseline phosphorylation levels of Src and ERK1/2 were drastically reduced. Both WT and A420P mutant hiPSC readily differentiated into cardiac myocytes (iCM), as evidenced by marker gene expression, spontaneous cell contraction, and subcellular striations. Total NKA α1-3 protein expression was comparable in WT and A420P iCM. However, live cell metabolism assessed functionally by Seahorse extracellular flux analysis revealed significant reductions in both basal and maximal rates of mitochondrial respiration, spare respiratory capacity, ATP production, and coupling efficiency. A significant reduction in ROS production was detected by fluorescence imaging in live cells, and confirmed by decreased cellular protein carbonylation levels in A420P iCM. Taken together, these data provide genetic evidence for a role of NKA α1/Src in the tonic stimulation of basal mitochondrial metabolism and ROS production in human cardiac myocytes. This signaling axis in cardiac myocytes may provide a new approach to counteract mitochondrial dysfunction in cardiometabolic diseases.
Collapse
Affiliation(s)
- Liquan Cai
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
| | - Marco T. Pessoa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
| | - Yingnyu Gao
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
| | - Sidney Strause
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
| | - Moumita Banerjee
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA
| | - Jiang Tian
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA; (L.C.); (M.T.P.); (Y.G.); (S.S.); (M.B.); (J.T.); (Z.X.)
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
31
|
Saha S, Fang X, Green CD, Das A. mTORC1 and SGLT2 Inhibitors-A Therapeutic Perspective for Diabetic Cardiomyopathy. Int J Mol Sci 2023; 24:15078. [PMID: 37894760 PMCID: PMC10606418 DOI: 10.3390/ijms242015078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetic cardiomyopathy is a critical diabetes-mediated co-morbidity characterized by cardiac dysfunction and heart failure, without predisposing hypertensive or atherosclerotic conditions. Metabolic insulin resistance, promoting hyperglycemia and hyperlipidemia, is the primary cause of diabetes-related disorders, but ambiguous tissue-specific insulin sensitivity has shed light on the importance of identifying a unified target paradigm for both the glycemic and non-glycemic context of type 2 diabetes (T2D). Several studies have indicated hyperactivation of the mammalian target of rapamycin (mTOR), specifically complex 1 (mTORC1), as a critical mediator of T2D pathophysiology by promoting insulin resistance, hyperlipidemia, inflammation, vasoconstriction, and stress. Moreover, mTORC1 inhibitors like rapamycin and their analogs have shown significant benefits in diabetes and related cardiac dysfunction. Recently, FDA-approved anti-hyperglycemic sodium-glucose co-transporter 2 inhibitors (SGLT2is) have gained therapeutic popularity for T2D and diabetic cardiomyopathy, even acknowledging the absence of SGLT2 channels in the heart. Recent studies have proposed SGLT2-independent drug mechanisms to ascertain their cardioprotective benefits by regulating sodium homeostasis and mimicking energy deprivation. In this review, we systematically discuss the role of mTORC1 as a unified, eminent target to treat T2D-mediated cardiac dysfunction and scrutinize whether SGLT2is can target mTORC1 signaling to benefit patients with diabetic cardiomyopathy. Further studies are warranted to establish the underlying cardioprotective mechanisms of SGLT2is under diabetic conditions, with selective inhibition of cardiac mTORC1 but the concomitant activation of mTORC2 (mTOR complex 2) signaling.
Collapse
Affiliation(s)
- Sumit Saha
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Xianjun Fang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Christopher D. Green
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
32
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
33
|
Sánchez-Gloria JL, Martínez-Olivares CE, Del Valle-Mondragón L, Cortés-Camacho F, Zambrano-Vásquez OR, Hernández-Pando R, Sánchez-Muñoz F, Sánchez-Lozada LG, Osorio-Alonso H. Allicin, an Emerging Treatment for Pulmonary Arterial Hypertension: An Experimental Study. Int J Mol Sci 2023; 24:12959. [PMID: 37629140 PMCID: PMC10454707 DOI: 10.3390/ijms241612959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
We assessed whether allicin, through its antihypertensive and antioxidant effects, relieves vascular remodeling, endothelial function, and oxidative stress (OS), thereby improving experimental pulmonary arterial hypertension (PAH). Allicin (16 mg/kg) was administered to rats with PAH (monocrotaline 60 mg/kg). Allicin encouraged body weight gain and survival rate, and medial wall thickness and the right ventricle (RV) hypertrophy were prevented. Also, angiotensin II concentrations in the lung (0.37 ± 0.01 vs. 0.47 ± 0.06 pmoles/mL, allicin and control, respectively) and plasma (0.57 ± 0.05 vs. 0.75 ± 0.064, allicin and control respectively) and the expressions of angiotensin-converting enzyme II and angiotensin II type 1 receptor in lung tissue were maintained at normal control levels with allicin. In PAH rats treated with allicin, nitric oxide (NO) (31.72 ± 1.22 and 51.4 ± 3.45 pmoles/mL), tetrahydrobiopterin (8.43 ± 0.33 and 10.14 ± 0.70 pmoles/mL), cyclic guanosine monophosphate (5.54 ± 0.42 and 5.64 ± 0.73 pmoles/mL), and Ang-(1-7) (0.88 ± 0.23 and 0.83 ± 0.056 pmoles/mL) concentrations increased in lung tissue and plasma, respectively. In contrast, dihydrobiopterin increase was prevented in both lung tissue and plasma (5.75 ± 0.3 and 5.64 ± 0.73 pmoles/mL); meanwhile, phosphodiesterase-5 was maintained at normal levels in lung tissue. OS in PAH was prevented with allicin through the increased expression of Nrf2 in the lung. Allicin prevented the lung response to hypoxia, preventing the overexpression of HIF-1α and VEGF. Allicin attenuated the vascular remodeling and RV hypertrophy in PAH through its effects on NO-dependent vasodilation, modulation of RAS, and amelioration of OS. Also, these effects could be associated with the modulation of HIF-1α and improved lung oxygenation. The global effects of allicin contribute to preventing endothelial dysfunction, remodeling of the pulmonary arteries, and RV hypertrophy, preventing heart failure, thus favoring survival. Although human studies are needed, the data suggest that, alone or in combination therapy, allicin may be an alternative in treating PAH if we consider that, similarly to current treatments, it improves lung vasodilation and increase survival. Allicin may be considered an option when there is a lack of efficacy, and where drug intolerance is observed, to enhance the efficacy of drugs, or when more than one pathogenic mechanism must be addressed.
Collapse
Affiliation(s)
- José L. Sánchez-Gloria
- Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Constanza E. Martínez-Olivares
- Experimental Pathology Department, Experimental Pathology Laboratory, Instituto Nacional de Ciencia Médicas y Nutrición “Salvador Zubirán”, Mexico City 14080, Mexico; (C.E.M.-O.); (R.H.-P.)
| | - Leonardo Del Valle-Mondragón
- Departamento de Farmacología “Dr. Rafael Méndez Martínez”, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Fernando Cortés-Camacho
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| | - Oscar R. Zambrano-Vásquez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| | - Rogelio Hernández-Pando
- Experimental Pathology Department, Experimental Pathology Laboratory, Instituto Nacional de Ciencia Médicas y Nutrición “Salvador Zubirán”, Mexico City 14080, Mexico; (C.E.M.-O.); (R.H.-P.)
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Laura G. Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| |
Collapse
|
34
|
Renaud D, Scholl-Bürgi S, Karall D, Michel M. Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy. Metabolites 2023; 13:932. [PMID: 37623876 PMCID: PMC10456471 DOI: 10.3390/metabo13080932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
Most studies on single ventricle (SV) circulation take a physiological or anatomical approach. Although there is a tight coupling between cardiac contractility and metabolism, the metabolic perspective on this patient population is very recent. Early findings point to major metabolic disturbances, with both impaired glucose and fatty acid oxidation in the cardiomyocytes. Additionally, Fontan patients have systemic metabolic derangements such as abnormal glucose metabolism and hypocholesterolemia. Our literature review compares the metabolism of patients with a SV circulation after Fontan palliation with that of patients with a healthy biventricular (BV) heart, or different subtypes of a failing BV heart, by Pubmed review of the literature on cardiac metabolism, Fontan failure, heart failure (HF), ketosis, metabolism published in English from 1939 to 2023. Early evidence demonstrates that SV circulation is not only a hemodynamic burden requiring staged palliation, but also a metabolic issue with alterations similar to what is known for HF in a BV circulation. Alterations of fatty acid and glucose oxidation were found, resulting in metabolic instability and impaired energy production. As reported for patients with BV HF, stimulating ketone oxidation may be an effective treatment strategy for HF in these patients. Few but promising clinical trials have been conducted thus far to evaluate therapeutic ketosis with HF using a variety of instruments, including ketogenic diet, ketone esters, and sodium-glucose co-transporter-2 (SGLT2) inhibitors. An initial trial on a small cohort demonstrated favorable outcomes for Fontan patients treated with SGLT2 inhibitors. Therapeutic ketosis is worth considering in the treatment of Fontan patients, as ketones positively affect not only the myocardial energy metabolism, but also the global Fontan physiopathology. Induced ketosis seems promising as a concerted therapeutic strategy.
Collapse
Affiliation(s)
- David Renaud
- Fundamental and Biomedical Sciences, Paris-Cité University, 75006 Paris, France
- Health Sciences Faculty, Universidad Europea Miguel de Cervantes, 47012 Valladolid, Spain
- Fundacja Recover, 05-124 Skrzeszew, Poland
| | - Sabine Scholl-Bürgi
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Daniela Karall
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Miriam Michel
- Department of Child and Adolescent Health, Division of Pediatrics III—Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
35
|
de Koning MSLY, Emmens JE, Romero-Hernández E, Bourgonje AR, Assa S, Figarska SM, Cleland JGF, Samani NJ, Ng LL, Lang CC, Metra M, Filippatos GS, van Veldhuisen DJ, Anker SD, Dickstein K, Voors AA, Lipsic E, van Goor H, van der Harst P. Systemic oxidative stress associates with disease severity and outcome in patients with new-onset or worsening heart failure. Clin Res Cardiol 2023; 112:1056-1066. [PMID: 36997667 PMCID: PMC10062262 DOI: 10.1007/s00392-023-02171-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/08/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND Oxidative stress may be a key pathophysiological mediator in the development and progression of heart failure (HF). The role of serum-free thiol concentrations, as a marker of systemic oxidative stress, in HF remains largely unknown. OBJECTIVE The purpose of this study was to investigate associations between serum-free thiol concentrations and disease severity and clinical outcome in patients with new-onset or worsening HF. METHODS Serum-free thiol concentrations were determined by colorimetric detection in 3802 patients from the BIOlogy Study to TAilored Treatment in Chronic Heart Failure (BIOSTAT-CHF). Associations between free thiol concentrations and clinical characteristics and outcomes, including all-cause mortality, cardiovascular mortality, and a composite of HF hospitalization and all-cause mortality during a 2-years follow-up, were reported. RESULTS Lower serum-free thiol concentrations were associated with more advanced HF, as indicated by worse NYHA class, higher plasma NT-proBNP (P < 0.001 for both) and with higher rates of all-cause mortality (hazard ratio (HR) per standard deviation (SD) decrease in free thiols: 1.253, 95% confidence interval (CI): 1.171-1.341, P < 0.001), cardiovascular mortality (HR per SD: 1.182, 95% CI: 1.086-1.288, P < 0.001), and the composite outcome (HR per SD: 1.058, 95% CI: 1.001-1.118, P = 0.046). CONCLUSIONS In patients with new-onset or worsening HF, a lower serum-free thiol concentration, indicative of higher oxidative stress, is associated with increased HF severity and poorer prognosis. Our results do not prove causality, but our findings may be used as rationale for future (mechanistic) studies on serum-free thiol modulation in heart failure. Associations of serum-free thiol concentrations with heart failure severity and outcomes.
Collapse
Affiliation(s)
- Marie-Sophie L Y de Koning
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands.
| | - Johanna E Emmens
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | | | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Solmaz Assa
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Sylwia M Figarska
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - John G F Cleland
- National Heart and Lung Institute, Royal Brompton and Harefield Hospitals, Imperial College, London, UK
- Robertson Centre for Biostatistics and Clinical Trials, University of Glasgow, Glasgow, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester, NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Chim C Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Marco Metra
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Institute of Cardiology, University of Brescia, Brescia, Italy
| | | | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Stefan D Anker
- Department of Cardiology (CVK), Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin Institute of Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Erik Lipsic
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB, Groningen, The Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
36
|
Yaluri N, Stančáková Yaluri A, Žeňuch P, Žeňuchová Z, Tóth Š, Kalanin P. Cardiac Biomarkers and Their Role in Identifying Increased Risk of Cardiovascular Complications in COVID-19 Patients. Diagnostics (Basel) 2023; 13:2508. [PMID: 37568870 PMCID: PMC10417576 DOI: 10.3390/diagnostics13152508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular disease (CVD) is a global health concern, causing significant morbidity and mortality. Both lifestyle and genetics influence the development of CVD. It is often diagnosed late, when the treatment options are limited. Early diagnosis of CVD with help of biomarkers is necessary to prevent adverse outcomes. SARS-CoV-2 infection can cause cardiovascular complications even in patients with no prior history of CVD. This review highlights cardiovascular biomarkers, including novel ones, and their applications as diagnostic and prognostic markers of cardiovascular complications related to SARS-CoV-2 infection. Patients with severe SARS-CoV-2 infection were shown to have elevated levels of cardiac biomarkers, namely N-terminal pro-brain natriuretic peptide (NT-pro-BNP), creatine kinase-myocardial band (CK-MB), and troponins, indicating acute myocardial damage. These biomarkers were also associated with higher mortality rates and therefore should be used throughout COVID-19 patient care to identify high-risk patients promptly to optimize their outcomes. Additionally, microRNAs (miRNAs) are also considered as potential biomarkers and predictors of cardiac and vascular damage in SARS-CoV-2 infection. Identifying molecular pathways contributing to cardiovascular manifestations in COVID-19 is essential for development of early biomarkers, identification of new therapeutic targets, and better prediction and management of cardiovascular outcomes.
Collapse
Affiliation(s)
- Nagendra Yaluri
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | | | - Pavol Žeňuch
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Zuzana Žeňuchová
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Štefan Tóth
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Peter Kalanin
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
37
|
Cohen-Segev R, Nativ O, Kinaneh S, Aronson D, Kabala A, Hamoud S, Karram T, Abassi Z. Effects of Angiotensin 1-7 and Mas Receptor Agonist on Renal System in a Rat Model of Heart Failure. Int J Mol Sci 2023; 24:11470. [PMID: 37511227 PMCID: PMC10380355 DOI: 10.3390/ijms241411470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Congestive heart failure (CHF) is often associated with impaired kidney function. Over- activation of the renin-angiotensin-aldosterone system (RAAS) contributes to avid salt/water retention and cardiac hypertrophy in CHF. While the deleterious effects of angiotensin II (Ang II) in CHF are well established, the biological actions of angiotensin 1-7 (Ang 1-7) are not fully characterized. In this study, we assessed the acute effects of Ang 1-7 (0.3, 3, 30 and 300 ng/kg/min, IV) on urinary flow (UF), urinary Na+ excretion (UNaV), glomerular filtration rate (GFR) and renal plasma flow )RPF) in rats with CHF induced by the placement of aortocaval fistula. Additionally, the chronic effects of Ang 1-7 (24 µg/kg/h, via intra-peritoneally implanted osmotic minipumps) on kidney function, cardiac hypertrophy and neurohormonal status were studied. Acute infusion of either Ang 1-7 or its agonist, AVE 0991, into sham controls, but not CHF rats, increased UF, UNaV, GFR, RPF and urinary cGMP. In the chronic protocols, untreated CHF rats displayed lower cumulative UF and UNaV than their sham controls. Chronic administration of Ang 1-7 and AVE 0991 exerted significant diuretic, natriuretic and kaliuretic effects in CHF rats, but not in sham controls. Serum creatinine and aldosterone levels were significantly higher in vehicle-treated CHF rats as compared with controls. Treatment with Ang 1-7 and AVE 0991 reduced these parameters to comparable levels observed in sham controls. Notably, chronic administration of Ang 1-7 to CHF rats reduced cardiac hypertrophy. In conclusion, Ang 1-7 exerts beneficial renal and cardiac effects in rats with CHF. Thus, we postulate that ACE2/Ang 1-7 axis represents a compensatory response to over-activity of ACE/AngII/AT1R system characterizing CHF and suggest that Ang 1-7 may be a potential therapeutic agent in this disease state.
Collapse
Affiliation(s)
- Ravit Cohen-Segev
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Omri Nativ
- Department of Urology, Rambam Health Center, Haifa 3109601, Israel
| | - Safa Kinaneh
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Doron Aronson
- Cardiology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Aviva Kabala
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Tony Karram
- Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
- Laboratory Medicine, Rambam Health Care Campus, Haifa 31096, Israel
| |
Collapse
|
38
|
Bazgir F, Nau J, Nakhaei-Rad S, Amin E, Wolf MJ, Saucerman JJ, Lorenz K, Ahmadian MR. The Microenvironment of the Pathogenesis of Cardiac Hypertrophy. Cells 2023; 12:1780. [PMID: 37443814 PMCID: PMC10341218 DOI: 10.3390/cells12131780] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Pathological cardiac hypertrophy is a key risk factor for the development of heart failure and predisposes individuals to cardiac arrhythmia and sudden death. While physiological cardiac hypertrophy is adaptive, hypertrophy resulting from conditions comprising hypertension, aortic stenosis, or genetic mutations, such as hypertrophic cardiomyopathy, is maladaptive. Here, we highlight the essential role and reciprocal interactions involving both cardiomyocytes and non-myocardial cells in response to pathological conditions. Prolonged cardiovascular stress causes cardiomyocytes and non-myocardial cells to enter an activated state releasing numerous pro-hypertrophic, pro-fibrotic, and pro-inflammatory mediators such as vasoactive hormones, growth factors, and cytokines, i.e., commencing signaling events that collectively cause cardiac hypertrophy. Fibrotic remodeling is mediated by cardiac fibroblasts as the central players, but also endothelial cells and resident and infiltrating immune cells enhance these processes. Many of these hypertrophic mediators are now being integrated into computational models that provide system-level insights and will help to translate our knowledge into new pharmacological targets. This perspective article summarizes the last decades' advances in cardiac hypertrophy research and discusses the herein-involved complex myocardial microenvironment and signaling components.
Collapse
Affiliation(s)
- Farhad Bazgir
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| | - Julia Nau
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| | - Saeideh Nakhaei-Rad
- Stem Cell Biology, and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad 91779-48974, Iran;
| | - Ehsan Amin
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Matthew J. Wolf
- Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA;
| | - Jeffry J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA;
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Leibniz Institute for Analytical Sciences, 97078 Würzburg, Germany;
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| |
Collapse
|
39
|
Stojanovic D, Stojanovic M, Milenkovic J, Velickov A, Ignjatovic A, Milojkovic M. The Multi-Faceted Nature of Renalase for Mitochondrial Dysfunction Improvement in Cardiac Disease. Cells 2023; 12:1607. [PMID: 37371077 DOI: 10.3390/cells12121607] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The cellular mechanisms and signaling network that guide the cardiac disease pathophysiology are inextricably intertwined, which explains the current scarcity of effective therapy and to date remains the greatest challenge in state-of-the-art cardiovascular medicine. Accordingly, a novel concept has emerged in which cardiomyocytes are the centerpiece of therapeutic targeting, with dysregulated mitochondria as a critical point of intervention. Mitochondrial dysfunction pluralism seeks a multi-faceted molecule, such as renalase, to simultaneously combat the pathophysiologic heterogeneity of mitochondria-induced cardiomyocyte injury. This review provides some original perspectives and, for the first time, discusses the functionality spectrum of renalase for mitochondrial dysfunction improvement within cardiac disease, including its ability to preserve mitochondrial integrity and dynamics by suppressing mitochondrial ΔΨm collapse; overall ATP content amelioration; a rise of mtDNA copy numbers; upregulation of mitochondrial genes involved in oxidative phosphorylation and cellular vitality promotion; mitochondrial fission inhibition; NAD+ supplementation; sirtuin upregulation; and anti-oxidant, anti-apoptotic, and anti-inflammatory traits. If verified that renalase, due to its multi-faceted nature, behaves like the "guardian of mitochondria" by thwarting pernicious mitochondrial dysfunction effects and exerting therapeutic potential to target mitochondrial abnormalities in failing hearts, it may provide large-scale benefits for cardiac disease patients, regardless of the underlying causes.
Collapse
Affiliation(s)
- Dijana Stojanovic
- Department of Pathophysiology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Miodrag Stojanovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
- Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, 18000 Nis, Serbia
| | - Jelena Milenkovic
- Department of Pathophysiology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Aleksandra Velickov
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Aleksandra Ignjatovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
- Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, 18000 Nis, Serbia
| | - Maja Milojkovic
- Department of Pathophysiology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| |
Collapse
|
40
|
Zahedi N, Pourajam S, Zaker E, Kouhpayeh S, Mirbod SM, Tavangar M, Boshtam M, Hatami Kahkesh K, Qian Q, Zhang F, Shariati L, Khanahmad H, Boshtam M. The potential therapeutic impacts of trehalose on cardiovascular diseases as the environmental-influenced disorders: An overview of contemporary findings. ENVIRONMENTAL RESEARCH 2023; 226:115674. [PMID: 36925035 DOI: 10.1016/j.envres.2023.115674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 06/18/2023]
Abstract
Cardiovascular diseases (CVDs) as environmental-influenced disorders, are a major concern and the leading cause of death worldwide. A range of therapeutic approaches has been proposed, including conventional and novel methods. Natural compounds offer a promising alternative for CVD treatment due to their ability to regulate molecular pathways with minimal adverse effects. Trehalose is natural compound and disaccharide with unique biological functions and cardio-protective properties. The cardio-protective effects of trehalose are generated through its ability to induce autophagy, which is mediated by the transcription factors TFEB and FOXO1. The stimulation of TFEB plays a significant role in regulating autophagy genes and autophagosome formation. Activation of FOXO1 through dephosphorylation of Foxo1 and blocking of p38 mitogen-activated protein kinase (p38 MAPK) also triggers autophagy dramatically. Trehalose has been shown to reduce CVD risk factors, including atherosclerosis, cardiac remodeling after a heart attack, cardiac dysfunction, high blood pressure, and stroke. It also reduces structural abnormalities of mitochondria, cytokine production, vascular inflammation, cardiomyocyte apoptosis, and pyroptosis. This review provides a molecular overview of trehalose's cardioprotective functions, including its mechanisms of autophagy and its potential to improve CVD symptoms based on clinical evidence.
Collapse
Affiliation(s)
- Noushin Zahedi
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samaneh Pourajam
- Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfan Zaker
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Shirin Kouhpayeh
- Department of Immunology, Erythron Genetics and Pathobiology Laboratory, Isfahan, Iran
| | - Seyedeh Mahnaz Mirbod
- Department of Cardiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrsa Tavangar
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Kaveh Hatami Kahkesh
- Department of Basic Medical Science, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Qiuping Qian
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, Zhejiang, China
| | - Feng Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, Zhejiang, China
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
41
|
Dutta A, Saha S, Bahl A, Mittal A, Basak T. A comprehensive review of acute cardio-renal syndrome: need for novel biomarkers. Front Pharmacol 2023; 14:1152055. [PMID: 37288107 PMCID: PMC10242013 DOI: 10.3389/fphar.2023.1152055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/03/2023] [Indexed: 06/09/2023] Open
Abstract
Cardiorenal syndrome represents a wide-spectrum disorder involving the heart and kidneys as the primary affected organs. India has an increasingly high burden of acute CRS, coinciding with the rise in global statistics. Up to 2022, approximately 46.1% of all cardiorenal patients have been diagnosed with acute CRS in India. Acute CRS involves a sudden deterioration of kidney functionalities, referred to as acute kidney injury (AKI) in acute heart failure patients. The pathophysiology of CRS involves hyperactivation of the sympathetic nervous system (SNS) and the renin-angiotensin-aldosterone system (RAAS) following acute myocardial stress. The pathological phenotype of acute CRS is associated with perturbed inflammatory, cellular, and neurohormonal markers in circulation. These complications increase the risk of mortality in clinically diagnosed acute CRS patients, making it a worldwide healthcare burden. Hence, effective diagnosis and early prevention are crucial to prevent the progression of CRS in AHF patients. Present biomarkers, such as serum creatinine (sCr), cystatin C (CysC), glomerular filtration rate (GFR), blood urea nitrogen (BUN), serum and/or urine neutrophil gelatinase-associated lipocalin (NGAL), B-type natriuretic peptide (BNP), and NT-proBNP, are clinically used to diagnose AKI stages in CRS patients but are limitedly sensitive to the early detection of the pathology. Therefore, the need for protein biomarkers is emerging for early intervention in CRS progression. Here, we summarized the cardio-renal nexus in acute CRS, with an emphasis on the present clinicopathological biomarkers and their limitations. The objective of this review is to highlight the need for novel proteomic biomarkers that will curb the burgeoning concern and direct future research trials.
Collapse
Affiliation(s)
- Abhi Dutta
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
| | - Shubham Saha
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
| | - Ajay Bahl
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anupam Mittal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
| |
Collapse
|
42
|
Martinez VR, Martins Lima A, Stergiopulos N, Velez Rueda JO, Islas MS, Griera M, Calleros L, Rodriguez Puyol M, Jaquenod de Giusti C, Portiansky EL, Ferrer EG, De Giusti V, Williams PAM. Effect of the structural modification of Candesartan with Zinc on hypertension and left ventricular hypertrophy. Eur J Pharmacol 2023; 946:175654. [PMID: 36930883 DOI: 10.1016/j.ejphar.2023.175654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/25/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Hypertension is the most common cause of left ventricular hypertrophy, contributing to heart failure progression. Candesartan (Cand) is an angiotensin receptor antagonist widely used for hypertension treatment. Structural modifications were previously performed by our group using Zinc (ZnCand) as a strategy for improving its pharmacological properties. The measurements showed that ZnCand exerts a stronger interaction with the angiotensin II receptor, type 1 (AT1 receptor), reducing oxidative stress and intracellular calcium flux, a mechanism implied in cell contraction. These results were accompanied by the reduction of the contractile capacity of mesangial cells. In vivo experiments showed that the complex causes a significant decrease in systolic blood pressure after 8 weeks of treatment in spontaneously hypertensive rats (SHR). The reduction of heart hypertrophy was evidenced by echocardiography, the histologic cross-sectional area of cardiomyocytes, collagen content, the B-type natriuretic peptide (BNP) marker and connective tissue growth factor (CTGF) and the matrix metalloproteinase 2 (MMP-2) expression. Besides, the complex restored the redox status. In this study, we demonstrated that the complexation with Zn(II) improves the antihypertensive and cardiac effects of the parental drug.
Collapse
Affiliation(s)
- Valeria R Martinez
- CEQUINOR-CONICET-CICPBA-UNLP, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv. 120 N◦ 1465, 1900, La Plata, Argentina; CIC-CONICET-UNLP, Facultad de Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina
| | - Augusto Martins Lima
- Laboratory of Hemodynamics & Cardiovascular Technology (LHTC), Institute of Bioengineering (Bâtiment MED), Station 9, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Nikolaous Stergiopulos
- Laboratory of Hemodynamics & Cardiovascular Technology (LHTC), Institute of Bioengineering (Bâtiment MED), Station 9, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Jorge O Velez Rueda
- CIC-CONICET-UNLP, Facultad de Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina
| | - Maria S Islas
- Departamento de Química y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, 7600, Mar del Plata, Argentina
| | - Mercedes Griera
- Departamento de Fisiología, Universidad de Alcalá, Campus Universitario, 28871, Alcalá de Henares, Madrid, Spain
| | - Laura Calleros
- Departamento de Fisiología, Universidad de Alcalá, Campus Universitario, 28871, Alcalá de Henares, Madrid, Spain
| | - Manuel Rodriguez Puyol
- Departamento de Fisiología, Universidad de Alcalá, Campus Universitario, 28871, Alcalá de Henares, Madrid, Spain
| | - Carolina Jaquenod de Giusti
- CIC-CONICET-UNLP, Facultad de Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina
| | - Enrique L Portiansky
- Laboratorio de Análisis de Imágenes-UNLP, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, 60 y 118, 1900, La Plata, Argentina
| | - Evelina G Ferrer
- CEQUINOR-CONICET-CICPBA-UNLP, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv. 120 N◦ 1465, 1900, La Plata, Argentina
| | - Verónica De Giusti
- CIC-CONICET-UNLP, Facultad de Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina.
| | - Patricia A M Williams
- CEQUINOR-CONICET-CICPBA-UNLP, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv. 120 N◦ 1465, 1900, La Plata, Argentina.
| |
Collapse
|
43
|
Rostamzadeh F, Jafarinejad-Farsangi S, Ansari-Asl Z, Farrokhi MS, Jafari E. Treatment for Myocardial Infarction: In Vivo Evaluation of Curcumin-Loaded PEGylated-GQD Nanoparticles. J Cardiovasc Pharmacol 2023; 81:361-372. [PMID: 36822208 DOI: 10.1097/fjc.0000000000001410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 02/04/2023] [Indexed: 02/25/2023]
Abstract
ABSTRACT Curcumin (Cur) has been suggested as a complementary treatment for cardiovascular diseases. Its efficiency, however, is modest due to poor biocompatibility. This study examined the effects of curcumin loaded on polyethylene glycol-graphene quantum dots (Cur-PEG-GQDs) on hemodynamic and cardiac function in rats with myocardial infarction (MI). The study groups included control, MI, MI+Cur-3, MI + Cur-7, MI + Cur-15, MI + PEG-GQDs-5, MI + PEG-GQDs-10, MI + Cur-PEG-GQDs-5, and MI + Cur-PEG-GQDs-10. MI was established by left anterior descending artery ligation. Two weeks after intraperitoneal administration of vehicle, Cur, PEG-GQDs, and Cur-PEG-GQDs, blood pressure and heart contractility indices were measured. Triphenyl tetrazolium chloride, colorimetry, and clinical laboratory methods were used to measure the infarct size, the oxidant and antioxidant content, and the kidney and liver function parameters, respectively. In the MI animals, Cur-7, PEG-GQDs-10, Cur-PEG-GQDs-5, and Cur-PEG-GQDs-10 recovered systolic blood pressure, diastolic blood pressure, left ventricular systolic pressure, and ±dp/dt max disturbances and reduced myocardial infarct size, fibrosis, and left ventricular end-diastolic pressure. Curcumin lowered antioxidant markers and elevated 1 oxidant marker in the heart in a dose-dependent manner. Although Cur-PEG-GQDs-5 and Cur-PEG-GQDs-10 reduced curcumin's oxidative stress effects, the superoxide dismutase, glutathione peroxidase, and total antioxidant capacity levels were significantly lower in Cur-PEG-GQDs-5 and Cur-PEG-GQDs-10 groups compared with the MI group. Malondialdehyde levels were lower in Cur-PEG-GQDs-5 and -10 groups compared with the Cur-3, Cur-7, and Cur-15 groups. The glutathione/glutathione disulfide ratio improved in the groups treated by Cur-7, PEG-GQDs-10, Cur-PEG-GQDs-5, and Cur-PEG-GQDs-10. The findings indicated that Cur-PEG-GQDs mitigated MI-induced cardiac dysfunction. However, because of the increase in oxidative stress in the heart, nonclassic mechanisms may be involved in the beneficial effect of Cur-PEG-GQDs on MI-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Farzaneh Rostamzadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman
| | - Saeideh Jafarinejad-Farsangi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman
| | - Zeinab Ansari-Asl
- Department of Chemistry, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz
| | - Mitra Shadkam Farrokhi
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman; and
| | - Elham Jafari
- Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
44
|
Lee SY, Kuo YH, Du CX, Huang CW, Ku HC. A novel caffeic acid derivative prevents angiotensin II-induced cardiac remodeling. Biomed Pharmacother 2023; 162:114709. [PMID: 37084559 DOI: 10.1016/j.biopha.2023.114709] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023] Open
Abstract
Differentiation of cardiac fibroblasts into myofibroblasts is a critical event in the progression of cardiac fibrosis that causes pathological cardiac remodeling. Cardiac fibrosis is a hallmark of heart disease and is associated with a stiff myocardium and heart failure. This study investigated the effect of caffeic acid ethanolamide (CAEA), a novel caffeic acid derivative, on cardiac remodeling. Angiotensin (Ang) II was used to induce cardiac remodeling both in cell and animal studies. Treating cardiac fibroblast with CAEA in Ang II-exposed cell cultures reduced the expression of fibrotic marker α-smooth muscle actin (α-SMA) and collagen and the production of superoxide, indicating that CAEA inhibited the differentiation of fibroblast into myofibroblast after Ang II exposure. CAEA protects against Ang II-induced cardiac fibrosis and dysfunction in vivo, characterized by the alleviation of collagen accumulation and the recovery of ejection fraction. In addition, CAEA decreased Ang II-induced transforming growth factor-β (TGF-β) expression and reduced NOX4 expression and oxidative stress in a SMAD-dependent pathway. CAEA participated in the regulation of Ang II-induced TGF-β/SMAD/NOX4 signaling to prevent the differentiation of fibroblast into myofibroblast and thus exerted a cardioprotective effect. Our data support the administration of CAEA as a viable method for preventing the progression of Ang II-induced cardiac remodeling.
Collapse
Affiliation(s)
- Shih-Yi Lee
- Division of Pulmonary and Critical Care Medicine, MacKay Memorial Hospital, Taipei, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Chen-Xuan Du
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Cheng-Wei Huang
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hui-Chun Ku
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
45
|
Maines E, Moretti M, Vitturi N, Gugelmo G, Fasan I, Lenzini L, Piccoli G, Gragnaniello V, Maiorana A, Soffiati M, Burlina A, Franceschi R. Understanding the Pathogenesis of Cardiac Complications in Patients with Propionic Acidemia and Exploring Therapeutic Alternatives for Those Who Are Not Eligible or Are Waiting for Liver Transplantation. Metabolites 2023; 13:563. [PMID: 37110221 PMCID: PMC10143878 DOI: 10.3390/metabo13040563] [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: 02/13/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The guidelines for the management of patients affected by propionic acidemia (PA) recommend standard cardiac therapy in the presence of cardiac complications. A recent revision questioned the impact of high doses of coenzyme Q10 on cardiac function in patients with cardiomyopathy (CM). Liver transplantation is a therapeutic option for several patients since it may stabilize or reverse CM. Both the patients waiting for liver transplantation and, even more, the ones not eligible for transplant programs urgently need therapies to improve cardiac function. To this aim, the identification of the pathogenetic mechanisms represents a key point. Aims: This review summarizes: (1) the current knowledge of the pathogenetic mechanisms underlying cardiac complications in PA and (2) the available and potential pharmacological options for the prevention or the treatment of cardiac complications in PA. To select articles, we searched the electronic database PubMed using the Mesh terms "propionic acidemia" OR "propionate" AND "cardiomyopathy" OR "Long QT syndrome". We selected 77 studies, enlightening 12 potential disease-specific or non-disease-specific pathogenetic mechanisms, namely: impaired substrate delivery to TCA cycle and TCA dysfunction, secondary mitochondrial electron transport chain dysfunction and oxidative stress, coenzyme Q10 deficiency, metabolic reprogramming, carnitine deficiency, cardiac excitation-contraction coupling alteration, genetics, epigenetics, microRNAs, micronutrients deficiencies, renin-angiotensin-aldosterone system activation, and increased sympathetic activation. We provide a critical discussion of the related therapeutic options. Current literature supports the involvement of multiple cellular pathways in cardiac complications of PA, indicating the growing complexity of their pathophysiology. Elucidating the mechanisms responsible for such abnormalities is essential to identify therapeutic strategies going beyond the correction of the enzymatic defect rather than engaging the dysregulated mechanisms. Although these approaches are not expected to be resolutive, they may improve the quality of life and slow the disease progression. Available pharmacological options are limited and tested in small cohorts. Indeed, a multicenter approach is mandatory to strengthen the efficacy of therapeutic options.
Collapse
Affiliation(s)
- Evelina Maines
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Michele Moretti
- Division of Cardiology, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Nicola Vitturi
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giorgia Gugelmo
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Ilaria Fasan
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Livia Lenzini
- Emergency Medicine Unit, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giovanni Piccoli
- CIBIO, Department of Cellular, Computational and Integrative Biology, Italy & Dulbecco Telethon Institute, Università degli Studi di Trento, 38123 Trento, Italy
| | - Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Arianna Maiorana
- Division of Metabolism and Research Unit of Metabolic Biochemistry, Bambino Gesù Children’s Hospital-IRCCS, 00165 Rome, Italy
| | - Massimo Soffiati
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Roberto Franceschi
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| |
Collapse
|
46
|
Chuaiphichai S, Chu SM, Carnicer R, Kelly M, Bendall JK, Simon JN, Douglas G, Crabtree MJ, Casadei B, Channon KM. Endothelial cell-specific roles for tetrahydrobiopterin in myocardial function, cardiac hypertrophy, and response to myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2023; 324:H430-H442. [PMID: 36735402 PMCID: PMC9988535 DOI: 10.1152/ajpheart.00562.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/15/2022] [Accepted: 12/31/2022] [Indexed: 02/04/2023]
Abstract
The cofactor tetrahydrobiopterin (BH4) is a critical regulator of nitric oxide synthase (NOS) function and redox signaling, with reduced BH4 implicated in multiple cardiovascular disease states. In the myocardium, augmentation of BH4 levels can impact on cardiomyocyte function, preventing hypertrophy and heart failure. However, the specific role of endothelial cell BH4 biosynthesis in the coronary circulation and its role in cardiac function and the response to ischemia has yet to be elucidated. Endothelial cell-specific Gch1 knockout mice were generated by crossing Gch1fl/fl with Tie2cre mice, generating Gch1fl/flTie2cre mice and littermate controls. GTP cyclohydrolase protein and BH4 levels were reduced in heart tissues from Gch1fl/flTie2cre mice, localized to endothelial cells, with normal cardiomyocyte BH4. Deficiency in coronary endothelial cell BH4 led to NOS uncoupling, decreased NO bioactivity, and increased superoxide and hydrogen peroxide productions in the hearts of Gch1fl/flTie2cre mice. Under physiological conditions, loss of endothelial cell-specific BH4 led to mild cardiac hypertrophy in Gch1fl/flTie2cre hearts. Endothelial cell BH4 loss was also associated with increased neuronal NOS protein, loss of endothelial NOS protein, and increased phospholamban phosphorylation at serine-17 in cardiomyocytes. Loss of cardiac endothelial cell BH4 led to coronary vascular dysfunction, reduced functional recovery, and increased myocardial infarct size following ischemia-reperfusion injury. Taken together, these studies reveal a specific role for endothelial cell Gch1/BH4 biosynthesis in cardiac function and the response to cardiac ischemia-reperfusion injury. Targeting endothelial cell Gch1 and BH4 biosynthesis may provide a novel therapeutic target for the prevention and treatment of cardiac dysfunction and ischemia-reperfusion injury.NEW & NOTEWORTHY We demonstrate a critical role for endothelial cell Gch1/BH4 biosynthesis in coronary vascular function and cardiac function. Loss of cardiac endothelial cell BH4 leads to coronary vascular dysfunction, reduced functional recovery, and increased myocardial infarct size following ischemia/reperfusion injury. Targeting endothelial cell Gch1 and BH4 biosynthesis may provide a novel therapeutic target for the prevention and treatment of cardiac dysfunction, ischemia injury, and heart failure.
Collapse
Affiliation(s)
- Surawee Chuaiphichai
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sandy M Chu
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matthew Kelly
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jenifer K Bendall
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jillian N Simon
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mark J Crabtree
- Department of Biochemical Sciences, School of Bioscience and Medicine, University of Surrey, Guildford, United Kingdom
| | - Barbara Casadei
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
47
|
Collins KB, Scott JD. Phosphorylation, compartmentalization, and cardiac function. IUBMB Life 2023; 75:353-369. [PMID: 36177749 PMCID: PMC10049969 DOI: 10.1002/iub.2677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022]
Abstract
Protein phosphorylation is a fundamental element of cell signaling. First discovered as a biochemical switch in glycogen metabolism, we now know that this posttranslational modification permeates all aspects of cellular behavior. In humans, over 540 protein kinases attach phosphate to acceptor amino acids, whereas around 160 phosphoprotein phosphatases remove phosphate to terminate signaling. Aberrant phosphorylation underlies disease, and kinase inhibitor drugs are increasingly used clinically as targeted therapies. Specificity in protein phosphorylation is achieved in part because kinases and phosphatases are spatially organized inside cells. A prototypic example is compartmentalization of the cyclic adenosine 3',5'-monophosphate (cAMP)-dependent protein kinase A through association with A-kinase anchoring proteins. This configuration creates autonomous signaling islands where the anchored kinase is constrained in proximity to activators, effectors, and selected substates. This article primarily focuses on A kinase anchoring protein (AKAP) signaling in the heart with an emphasis on anchoring proteins that spatiotemporally coordinate excitation-contraction coupling and hypertrophic responses.
Collapse
Affiliation(s)
- Kerrie B. Collins
- Department of Pharmacology, University of Washington, School of Medicine, 1959 NE Pacific Ave, Seattle WA, 98195
| | - John D. Scott
- Department of Pharmacology, University of Washington, School of Medicine, 1959 NE Pacific Ave, Seattle WA, 98195
| |
Collapse
|
48
|
Samir R, Hassan EA, Saber AA, Haneen DSA, Saleh EM. Seaweed Sargassum aquifolium extract ameliorates cardiotoxicity induced by doxorubicin in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:58226-58242. [PMID: 36977879 PMCID: PMC10163098 DOI: 10.1007/s11356-023-26259-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 02/28/2023] [Indexed: 05/08/2023]
Abstract
Doxorubicin (DOX) is a potent anticancer drug with adverse cardiotoxic effects. Alginates are multifunctional biopolymers and polyelectrolytes derived from the cell walls of brown seaweeds. They are nontoxic, biocompatible, and biodegradable, and hence, utilized in several biomedical and pharmaceutical applications. Here, we investigated the potential cardioprotective effect of thermally treated sodium alginate (TTSA), which was extracted and purified from the seaweed Sargassum aquifolium, in treating acute DOX cardiotoxicity and apoptotic pathways in rats. UV-visible spectroscopy, Fourier-transform infrared, and nuclear magnetic resonance (1H-NMR) spectroscopy techniques were used to characterize TTSA. CK-MB and AST levels in sera samples were determined. The expression levels of Erk-2 (MAPK-1) and iNOS genes were investigated by quantitative real-time polymerase chain reaction (qRT-PCR). The protein expression levels of Erk-2, anti-apoptotic p53, and caspase-3 were analyzed using western blotting and ELISA. For the in vivo studies, sixty rats were randomly divided equally into six groups and treated with DOX, followed by TTSA. We revealed that treatment with TTSA, which has low molecular weight and enhanced antioxidant properties, improved DOX-mediated cardiac dysfunction and alleviated DOX-induced myocardial apoptosis. Furthermore, TTSA exhibited a cardioprotective effect against DOX-induced cardiac toxicity, indicated by the increased expression of MAPK-1 (Erk2) and iNOS genes, which are implicated in the adaptive responses regulating DOX-induced myocardial damage. Moreover, TTSA significantly (p < 0.05) suppressed caspase-3 and upregulated anti-apoptotic protein p53 expression. TTSA also rebalanced the cardiomyocyte redox potential by significantly (p < 0.05) increasing the levels of endogenous antioxidant enzymes, including catalase and superoxide dismutase. Our findings suggest that TTSA, particularly at a dose of 400 mg/kg b.w., is a potential prophylactic supplement for treating acute DOX-linked cardiotoxicity.
Collapse
Affiliation(s)
- Rania Samir
- Biochemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| | - Ekrami A. Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| | - Abdullah A. Saber
- Botany Department, Faculty of Science, Ain Shams University, Abbassia Square, Cairo, 11566 Egypt
| | - David S. A. Haneen
- Chemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| | - Eman M. Saleh
- Biochemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| |
Collapse
|
49
|
Sasset L, Manzo OL, Zhang Y, Marino A, Rubinelli L, Riemma MA, Chalasani MLS, Dasoveanu DC, Roviezzo F, Jankauskas SS, Santulli G, Bucci MR, Lu TT, Di Lorenzo A. Nogo-A reduces ceramide de novo biosynthesis to protect from heart failure. Cardiovasc Res 2023; 119:506-519. [PMID: 35815623 PMCID: PMC10226746 DOI: 10.1093/cvr/cvac108] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Growing evidence correlate the accrual of the sphingolipid ceramide in plasma and cardiac tissue with heart failure (HF). Regulation of sphingolipid metabolism in the heart and the pathological impact of its derangement remain poorly understood. Recently, we discovered that Nogo-B, a membrane protein of endoplasmic reticulum, abundant in the vascular wall, down-regulates the sphingolipid de novo biosynthesis via serine palmitoyltransferase (SPT), first and rate liming enzyme, to impact vascular functions and blood pressure. Nogo-A, a splice isoform of Nogo, is transiently expressed in cardiomyocyte (CM) following pressure overload. Cardiac Nogo is up-regulated in dilated and ischaemic cardiomyopathies in animals and humans. However, its biological function in the heart remains unknown. METHODS AND RESULTS We discovered that Nogo-A is a negative regulator of SPT activity and refrains ceramide de novo biosynthesis in CM exposed to haemodynamic stress, hence limiting ceramide accrual. At 7 days following transverse aortic constriction (TAC), SPT activity was significantly up-regulated in CM lacking Nogo-A and correlated with ceramide accrual, particularly very long-chain ceramides, which are the most abundant in CM, resulting in the suppression of 'beneficial' autophagy. At 3 months post-TAC, mice lacking Nogo-A in CM showed worse pathological cardiac hypertrophy and dysfunction, with ca. 50% mortality rate. CONCLUSION Mechanistically, Nogo-A refrains ceramides from accrual, therefore preserves the 'beneficial' autophagy, mitochondrial function, and metabolic gene expression, limiting the progression to HF under sustained stress.
Collapse
Affiliation(s)
- Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Yi Zhang
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi 710061, China
| | - Alice Marino
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Luisa Rubinelli
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Maria Antonietta Riemma
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Madhavi Latha S Chalasani
- Department of Microbiology and Immunology, Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dragos C Dasoveanu
- Department of Microbiology and Immunology, Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Fiorentina Roviezzo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Stanislovas S Jankauskas
- Department of Medicine (Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Santulli
- Department of Medicine (Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Maria Rosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Theresa T Lu
- Department of Microbiology and Immunology, Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
50
|
Inhibition of Pyruvate Dehydrogenase in the Heart as an Initiating Event in the Development of Diabetic Cardiomyopathy. Antioxidants (Basel) 2023; 12:antiox12030756. [PMID: 36979003 PMCID: PMC10045649 DOI: 10.3390/antiox12030756] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Obesity affects a growing fraction of the population and is a risk factor for type 2 diabetes and cardiovascular disease. Even in the absence of hypertension and coronary artery disease, type 2 diabetes can result in a heart disease termed diabetic cardiomyopathy. Diminished glucose oxidation, increased reliance on fatty acid oxidation for energy production, and oxidative stress are believed to play causal roles. However, the progression of metabolic changes and mechanisms by which these changes impact the heart have not been established. Cardiac pyruvate dehydrogenase (PDH), the central regulatory site for glucose oxidation, is rapidly inhibited in mice fed high dietary fat, a model of obesity and diabetes. Increased reliance on fatty acid oxidation for energy production, in turn, enhances mitochondrial pro-oxidant production. Inhibition of PDH may therefore initiate metabolic inflexibility and oxidative stress and precipitate diabetic cardiomyopathy. We discuss evidence from the literature that supports a role for PDH inhibition in loss in energy homeostasis and diastolic function in obese and diabetic humans and in rodent models. Finally, seemingly contradictory findings highlight the complexity of the disease and the need to delineate progressive changes in cardiac metabolism, the impact on myocardial structure and function, and the ability to intercede.
Collapse
|