1
|
Ogilvie LM, Coyle-Asbil B, Brunt KR, Petrik J, Simpson JA. Therapy-naïve malignancy causes cardiovascular disease: a state-of-the-art cardio-oncology perspective. Am J Physiol Heart Circ Physiol 2024; 326:H1515-H1537. [PMID: 38639740 DOI: 10.1152/ajpheart.00795.2023] [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: 12/22/2023] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Cardiovascular disease (CVD) and cancer are the leading causes of mortality worldwide. Although generally thought of as distinct diseases, the intersectional overlap between CVD and cancer is increasingly evident in both causal and mechanistic relationships. The field of cardio-oncology is largely focused on the cardiotoxic effects of cancer therapies (e.g., chemotherapy, radiation). Furthermore, the cumulative effects of cardiotoxic therapy exposure and the prevalence of CVD risk factors in patients with cancer lead to long-term morbidity and poor quality of life in this patient population, even when patients are cancer-free. Evidence from patients with cancer and animal models demonstrates that the presence of malignancy itself, independent of cardiotoxic therapy exposure or CVD risk factors, negatively impacts cardiac structure and function. As such, the primary focus of this review is the cardiac pathophysiological and molecular features of therapy-naïve cancer. We also summarize the strengths and limitations of preclinical cancer models for cardio-oncology research and discuss therapeutic strategies that have been tested experimentally for the treatment of cancer-induced cardiac atrophy and dysfunction. Finally, we explore an adjacent area of interest, called "reverse cardio-oncology," where the sequelae of heart failure augment cancer progression. Here, we emphasize the cross-disease communication between malignancy and the injured heart and discuss the importance of chronic low-grade inflammation and endocrine factors in the progression of both diseases.
Collapse
Affiliation(s)
- Leslie M Ogilvie
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Bridget Coyle-Asbil
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Keith R Brunt
- Department of Pharmacology, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
- IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
- IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| |
Collapse
|
2
|
Ma Y, Zhao HP, Yang LG, Li L, Wang AL, Zhang XJ, Wang K, Yang B, Zhu ZF, Zhang PJ, Wang JP, Chi RF, Li B, Qin FZ, Wang ZP. NADPH oxidase 2 mediates cardiac sympathetic denervation and myocyte autophagy, resulting in cardiac atrophy and dysfunction in doxorubicin-induced cardiomyopathy. Sci Rep 2024; 14:6971. [PMID: 38521855 PMCID: PMC10960835 DOI: 10.1038/s41598-024-57090-2] [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: 07/01/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Doxorubicin has been used extensively as a potent anticancer agent, but its clinical use is limited by its cardiotoxicity. However, the underlying mechanisms remain to be fully elucidated. In this study, we tested whether NADPH oxidase 2 (Nox2) mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, resulting in cardiac atrophy and dysfunction in doxorubicin-induced heart failure. Nox2 knockout (KO) and wild-type (WT) mice were randomly assigned to receive a single injection of doxorubicin (15 mg/kg, i.p.) or saline. WT doxorubicin mice exhibited the decreases in survival rate, left ventricular (LV) wall thickness and LV fractional shortening and the increase in the lung wet-to-dry weight ratio 1 week after the injections. These alterations were attenuated in Nox2 KO doxorubicin mice. In WT doxorubicin mice, myocardial oxidative stress was increased, myocardial noradrenergic nerve fibers were reduced, myocardial expression of PGP9.5, GAP43, tyrosine hydroxylase and norepinephrine transporter was decreased, and these changes were prevented in Nox2 KO doxorubicin mice. Myocyte autophagy was increased and myocyte size was decreased in WT doxorubicin mice, but not in Nox2 KO doxorubicin mice. Nox2 mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy-both of which contribute to cardiac atrophy and failure after doxorubicin treatment.
Collapse
Affiliation(s)
- Yuan Ma
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Hui-Ping Zhao
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Li-Guo Yang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Lu Li
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Ai-Lin Wang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Xiao-Juan Zhang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Ke Wang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Bin Yang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Zong-Feng Zhu
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Pei-Jun Zhang
- Shanxi Datong University School of Medicine, Datong, 037009, Shanxi, People's Republic of China
| | - Jia-Pu Wang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Rui-Fang Chi
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Bao Li
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Fu-Zhong Qin
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China.
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China.
| | - Zhi-Peng Wang
- Institute for Radiation Protection, Taiyuan, 030006, Shanxi, People's Republic of China
| |
Collapse
|
3
|
Zhao HP, Ma Y, Zhang XJ, Guo HX, Yang B, Chi RF, Zhang NP, Wang JP, Li B, Qin FZ, Yang LG. NADPH oxidase 2 inhibitor GSK2795039 prevents doxorubicin-induced cardiac atrophy by attenuating cardiac sympathetic nerve terminal abnormalities and myocyte autophagy. Eur J Pharmacol 2024; 967:176351. [PMID: 38290568 DOI: 10.1016/j.ejphar.2024.176351] [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: 09/08/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
Doxorubicin is widely used for the treatment of human cancer, but its clinical use is limited by a cumulative dose-dependent cardiotoxicity. However, the mechanism of doxorubicin-induced cardiac atrophy and failure remains to be fully understood. In this study, we tested whether the specific NADPH oxidase 2 (Nox2) inhibitor GSK2795039 attenuates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, leading to the amelioration of cardiac atrophy and dysfunction in chronic doxorubicin-induced cardiomyopathy. Mice were randomized to receive saline, doxorubicin (2.5 mg/kg, every other day, 6 times) or doxorubicin plus GSK2795039 (2.5 mg/kg, twice a day, 9 weeks). Left ventricular (LV) total wall thickness and LV ejection fraction were decreased in doxorubicin-treated mice compared with saline-treated mice and the decreases were prevented by the treatment of the specific Nox2 inhibitor GSK2795039. The ratio of total heart weight to tibia length and myocyte cross-sectional area were decreased in doxorubicin-treated mice, and the decreases were attenuated by the GSK2795039 treatment. In doxorubicin-treated mice, myocardial Nox2 and 4-hydroxynonenal levels were increased, myocardial expression of GAP43, tyrosine hydroxylase and norepinephrine transporter, markers of sympathetic nerve terminals, was decreased, and these changes were prevented by the GSK2795039 treatment. The ratio of LC3 II/I, a marker of autophagy, and Atg5, Atg12 and Atg12-Atg5 conjugate proteins were increased in doxorubicin-treated mice, and the increases were attenuated by the GSK2795039 treatment. These findings suggest that inhibition of Nox2 by GSK2795039 attenuates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, thereby ameliorating cardiac atrophy and dysfunction after chronic doxorubicin treatment.
Collapse
Affiliation(s)
- Hui-Ping Zhao
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Yuan Ma
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Xiao-Juan Zhang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Hong-Xia Guo
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Bin Yang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Rui-Fang Chi
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Nian-Ping Zhang
- Shanxi Datong University School of Medicine, Datong, 037009, Shanxi, PR China
| | - Jia-Pu Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Bao Li
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Fu-Zhong Qin
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China.
| | - Li-Guo Yang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Provincial People's Hospital, Taiyuan, 030001, Shanxi, PR China
| |
Collapse
|
4
|
Bacova BS, Andelova K, Sykora M, Egan Benova T, Barancik M, Kurahara LH, Tribulova N. Does Myocardial Atrophy Represent Anti-Arrhythmic Phenotype? Biomedicines 2022; 10:2819. [PMID: 36359339 PMCID: PMC9687767 DOI: 10.3390/biomedicines10112819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2023] Open
Abstract
This review focuses on cardiac atrophy resulting from mechanical or metabolic unloading due to various conditions, describing some mechanisms and discussing possible strategies or interventions to prevent, attenuate or reverse myocardial atrophy. An improved awareness of these conditions and an increased focus on the identification of mechanisms and therapeutic targets may facilitate the development of the effective treatment or reversion for cardiac atrophy. It appears that a decrement in the left ventricular mass itself may be the central component in cardiac deconditioning, which avoids the occurrence of life-threatening arrhythmias. The depressed myocardial contractility of atrophied myocardium along with the upregulation of electrical coupling protein, connexin43, the maintenance of its topology, and enhanced PKCƐ signalling may be involved in the anti-arrhythmic phenotype. Meanwhile, persistent myocardial atrophy accompanied by oxidative stress and inflammation, as well as extracellular matrix fibrosis, may lead to severe cardiac dysfunction, and heart failure. Data in the literature suggest that the prevention of heart failure via the attenuation or reversion of myocardial atrophy is possible, although this requires further research.
Collapse
Affiliation(s)
| | - Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Tamara Egan Benova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Miroslav Barancik
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Lin Hai Kurahara
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Japan
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| |
Collapse
|
5
|
Germain P, Delalande A, Pichon C. Role of Muscle LIM Protein in Mechanotransduction Process. Int J Mol Sci 2022; 23:ijms23179785. [PMID: 36077180 PMCID: PMC9456170 DOI: 10.3390/ijms23179785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/14/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
The induction of protein synthesis is crucial to counteract the deconditioning of neuromuscular system and its atrophy. In the past, hormones and cytokines acting as growth factors involved in the intracellular events of these processes have been identified, while the implications of signaling pathways associated with the anabolism/catabolism ratio in reference to the molecular mechanism of skeletal muscle hypertrophy have been recently identified. Among them, the mechanotransduction resulting from a mechanical stress applied to the cell appears increasingly interesting as a potential pathway for therapeutic intervention. At present, there is an open question regarding the type of stress to apply in order to induce anabolic events or the type of mechanical strain with respect to the possible mechanosensing and mechanotransduction processes involved in muscle cells protein synthesis. This review is focused on the muscle LIM protein (MLP), a structural and mechanosensing protein with a LIM domain, which is expressed in the sarcomere and costamere of striated muscle cells. It acts as a transcriptional cofactor during cell proliferation after its nuclear translocation during the anabolic process of differentiation and rebuilding. Moreover, we discuss the possible opportunity of stimulating this mechanotransduction process to counteract the muscle atrophy induced by anabolic versus catabolic disorders coming from the environment, aging or myopathies.
Collapse
Affiliation(s)
- Philippe Germain
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Anthony Delalande
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Chantal Pichon
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
- Institut Universitaire de France, 1 Rue Descartes, 75231 Paris, France
- Correspondence:
| |
Collapse
|
6
|
Abstract
Sarcopenia is common in aging and in patients with heart failure (HF) who may experience worse outcomes. Patients with muscle wasting are more likely to experience falls and can have serious complications when undergoing cardiac procedures. While intensive nutritional support and exercise rehabilitation can help reverse some of these changes, they are often under-prescribed in a timely manner, and we have limited insights into who would benefit. Mechanistic links between gut microbial metabolites (GMM) have been identified and may contribute to adverse clinical outcomes in patients with cardio-renal diseases and aging. This review will examine the emerging evidence for the influence of the gut microbiome-derived metabolites and notable signaling pathways involved in both sarcopenia and HF, especially those linked to dietary intake and mitochondrial metabolism. This provides a unique opportunity to gain mechanistic and clinical insights into developing novel therapeutic strategies that target these GMM pathways or through tailored nutritional modulation to prevent progressive muscle wasting in elderly patients with heart failure.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, OH 44195, USA
| | - W H Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, OH 44195, USA.,Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
7
|
Dhanyalayam D, Thangavel H, Lizardo K, Oswal N, Dolgov E, Perlin DS, Nagajyothi JF. Sex Differences in Cardiac Pathology of SARS-CoV2 Infected and Trypanosoma cruzi Co-infected Mice. Front Cardiovasc Med 2022; 9:783974. [PMID: 35369283 PMCID: PMC8965705 DOI: 10.3389/fcvm.2022.783974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/11/2022] [Indexed: 12/03/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2; CoV2) is a deadly contagious infectious disease. For those who survive COVID-19, post-COVID cardiac damage greatly increases the risk of cardiomyopathy and heart failure. Currently, the number of COVID-related cases are increasing in Latin America, where a major COVID comorbidity is Chagas' heart disease, which is caused by the parasite Trypanosoma cruzi. However, the interplay between indeterminate Chagas disease and COVID-19 is unknown. We investigated the effect of CoV2 infection on heart pathology in T. cruzi infected mice (coinfected with CoV2 during the indeterminate stage of T. cruzi infection). We used transgenic human angiotensin-converting enzyme 2 (huACE2/hACE2) mice infected with CoV2, T. cruzi, or coinfected with both in this study. We found that the viral load in the hearts of coinfected mice is lower compared to the hearts of mice infected with CoV2 alone. We demonstrated that CoV2 infection significantly alters cardiac immune and energy signaling via adiponectin (C-ApN) and AMP-activated protein kinase (AMPK) signaling. Our studies also showed that increased β-adrenergic receptor (b-AR) and peroxisome proliferator-activated receptors (PPARs) play a major role in shifting the energy balance in the hearts of coinfected female mice from glycolysis to mitochondrial β-oxidation. Our findings suggest that cardiac metabolic signaling may differently regulate the pathogenesis of Chagas cardiomyopathy (CCM) in coinfected mice. We conclude that the C-ApN/AMPK and b-AR/PPAR downstream signaling may play major roles in determining the progression, severity, and phenotype of CCM and heart failure in the context of COVID.
Collapse
|
8
|
Cardiac Complications: The Understudied Aspect of Cancer Cachexia. Cardiovasc Toxicol 2022; 22:254-267. [PMID: 35171467 DOI: 10.1007/s12012-022-09727-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/03/2022] [Indexed: 12/17/2022]
Abstract
The global burden of cancer cachexia is increasing along with drastic increase in cancer patients. Cancer itself leads to cachexia, and cachexia development is associated with events like altered hemodynamics, and reduced functional capacity of the heart among others which lead to failure of the heart and are called cardiovascular complications associated with cancer cachexia. In some patients, the anti-cancer therapy also leads to this cardiovascular complications. So, in this review, an attempt is made to understand the mechanisms, pathophysiology of cardiovascular events in cachectic patients. Important processes which cause cardiovascular complications include alterations in the structure of the heart, loss of cardiac mass and functioning, cardiac fibrosis and cardiac remodeling, apoptosis, cardiac muscle atrophy, and mitochondrial alterations. Previously, the available treatment options were limited to nutraceuticals and physical exercise. Recently, studies with some prospective agents that can improve cardiac health have been reported, but whether their action is effective in cardiovascular complications associated with cancer cachexia is not known or are under trial.
Collapse
|
9
|
Gillis C, Ljungqvist O, Carli F. Prehabilitation, enhanced recovery after surgery, or both? A narrative review. Br J Anaesth 2022; 128:434-448. [PMID: 35012741 DOI: 10.1016/j.bja.2021.12.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/03/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
This narrative review presents a biological rationale and evidence to describe how the preoperative condition of the patient contributes to postoperative morbidity. Any preoperative condition that prevents a patient from tolerating the physiological stress of surgery (e.g. poor cardiopulmonary reserve, sarcopaenia), impairs the stress response (e.g. malnutrition, frailty), and/or augments the catabolic response to stress (e.g. insulin resistance) is a risk factor for poor surgical outcomes. Prehabilitation interventions that include exercise, nutrition, and psychosocial components can be applied before surgery to strengthen physiological reserve and enhance functional capacity, which, in turn, supports recovery through attaining surgical resilience. Prehabilitation complements Enhanced Recovery After Surgery (ERAS) care to achieve optimal patient outcomes because recovery is not a passive process and it begins preoperatively.
Collapse
Affiliation(s)
- Chelsia Gillis
- Department of Anesthesia, McGill University Health Center, Montreal, QC, Canada.
| | - Olle Ljungqvist
- Faculty of Medicine and Health, School of Health and Medical Sciences, Department of Surgery, Örebro University, Örebro, Sweden
| | - Francesco Carli
- Department of Anesthesia, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
10
|
Function and regulation of phosphatase 1 in healthy and diseased heart. Cell Signal 2021; 90:110203. [PMID: 34822978 DOI: 10.1016/j.cellsig.2021.110203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022]
Abstract
Reversible phosphorylation of ion channels and calcium-handling proteins provides precise post-translational regulation of cardiac excitation and contractility. Serine/threonine phosphatases govern dephosphorylation of the majority of cardiac proteins. Accordingly, dysfunction of this regulation contributes to the development and progression of heart failure and atrial fibrillation. On the molecular level, these changes include alterations in the expression level and phosphorylation status of Ca2+ handling and excitation-contraction coupling proteins provoked by dysregulation of phosphatases. The serine/threonine protein phosphatase PP1 is one a major player in the regulation of cardiac excitation-contraction coupling. PP1 essentially impacts on cardiac physiology and pathophysiology via interactions with the cardiac ion channels Cav1.2, NKA, NCX and KCNQ1, sarcoplasmic reticulum-bound Ca2+ handling proteins such as RyR2, SERCA and PLB as well as the contractile proteins MLC2, TnI and MyBP-C. PP1 itself but also PP1-regulatory proteins like inhibitor-1, inhibitor-2 and heat-shock protein 20 are dysregulated in cardiac disease. Therefore, they represent interesting targets to gain more insights in heart pathophysiology and to identify new treatment strategies for patients with heart failure or atrial fibrillation. We describe the genetic and holoenzymatic structure of PP1 and review its role in the heart and cardiac disease. Finally, we highlight the importance of the PP1 regulatory proteins for disease manifestation, provide an overview of genetic models to study the role of PP1 for the development of heart failure and atrial fibrillation and discuss possibilities of pharmacological interventions.
Collapse
|
11
|
Anthracycline-free tumor elimination in mice leads to functional and molecular cardiac recovery from cancer-induced alterations in contrast to long-lasting doxorubicin treatment effects. Basic Res Cardiol 2021; 116:61. [PMID: 34669013 PMCID: PMC8528750 DOI: 10.1007/s00395-021-00902-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 11/03/2022]
Abstract
Systemic effects of advanced cancer impact on the heart leading to cardiac atrophy and functional impairment. Using a murine melanoma cancer model (B16F10 melanoma cells stably transduced with a Ganciclovir (GCV)-inducible suicide gene), the present study analysed the recovery potential of cancer-induced cardiomyopathy with or without use of doxorubicin (Dox). After Dox-free tumor elimination and recovery for 70 ± 5 days, cancer-induced morphologic, functional, metabolic and molecular changes were largely reversible in mice previously bearing tumors. Moreover, grip strength and cardiac response to angiotensin II-induced high blood pressure were comparable with healthy control mice. In turn, addition of Dox (12 mg/kg BW) to melanoma-bearing mice reduced survival in the acute phase compared to GCV-alone induced recovery, while long-term effects on cardiac morphologic and functional recovery were similar. However, Dox treatment was associated with permanent changes in the cardiac gene expression pattern, especially the circadian rhythm pathway associated with the DNA damage repair system. Thus, the heart can recover from cancer-induced damage after chemotherapy-free tumor elimination. In contrast, treatment with the cardiotoxic drug Dox induces, besides well-known adverse acute effects, long-term subclinical changes in the heart, especially of circadian clock genes. Since the circadian clock is known to impact on cardiac repair mechanisms, these changes may render the heart more sensitive to additional stress during lifetime, which, at least in part, could contribute to late cardiac toxicity.
Collapse
|
12
|
Tourki B, Halade GV. Heart Failure Syndrome With Preserved Ejection Fraction Is a Metabolic Cluster of Non-resolving Inflammation in Obesity. Front Cardiovasc Med 2021; 8:695952. [PMID: 34409075 PMCID: PMC8367012 DOI: 10.3389/fcvm.2021.695952] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an emerging disease with signs of nonresolving inflammation, endothelial dysfunction, and multiorgan defects. Moreover, based on the clinical signs and symptoms and the rise of the obesity epidemic, the number of patients developing HFpEF is increasing. From recent molecular and cellular studies, it becomes evident that HFpEF is not a single and homogenous disease but a cluster of heterogeneous pathophysiology with aging at the base of the pyramid. Obesity superimposed on aging drives the number of inflammatory pathways that intersect with metabolic dysfunction and suboptimal inflammation. Here, we compiled information on obesity-directed macrophage dysfunction that coincide with metabolic defects. Obesity-associated proinflammatory stimuli facilitates heart and interorgan inflammation in HFpEF. Furthermore, diversified mechanisms that drive heart failure urge the need of studying pervasive and unresolved inflammation in animal models to understand HFpEF. A broad and system-based approach will help to study major translational aspects of HFpEF, since no single animal model recapitulates all signs of differential HFpEF stages in the clinical setting. Here, we covered experimental models that target HFpEF and emphasized the advances observed with formyl peptide 2 (FPR2) receptor, a prime sensor that is important in inflammation-resolution signaling. Dysfunction of FPR2 led to the development of spontaneous obesity, impaired macrophage function, and triggered kidney fibrosis, providing evidence of multiorgan defects in HFpEF in an obesogenic aging experimental model.
Collapse
Affiliation(s)
- Bochra Tourki
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| |
Collapse
|
13
|
Goda AE, Elenany AM, Elsisi AE. Novel in vivo potential of trifluoperazine to ameliorate doxorubicin-induced cardiotoxicity involves suppression of NF-κB and apoptosis. Life Sci 2021; 283:119849. [PMID: 34343539 DOI: 10.1016/j.lfs.2021.119849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/13/2021] [Accepted: 07/17/2021] [Indexed: 11/25/2022]
Abstract
AIMS Cardiotoxicity of doxorubicin frequently complicates treatment outcome. Aberrantly activated calcium/calmodulin pathway can eventually trigger signaling cascades that mediate cardiotoxicity. Therefore, we tested the hypothesis that trifluoperazine, a strong calmodulin antagonist, may alleviate this morbidity. MATERIALS AND METHODS Heart failure and cardiotoxicity were assessed via echocardiography, PCR, immunohistochemistry, histopathology, Masson's trichrome staining and transmission electron microscopy. Whereas liver and kidney structural and functional alterations were evaluated histopathologically and biochemically. KEY FINDINGS Results revealed that combination treatment with trifluoperazine could overcome doxorubicin-induced heart failure with reduced ejection fraction. Moreover, heart weight/body weight ratio and histopathological examination showed that trifluoperazine mitigated doxorubicin-induced cardiac atrophy, inflammation and myofibril degeneration. Transmission electron microscopy further confirmed the marked restoration of the left ventricular ultrastructures by trifluoperazine pretreatment. In addition, Masson's trichrome staining revealed that trifluoperazine could significantly inhibit doxorubicin-induced left ventricular remodeling by fibrosis. Of note, doxorubicin induced the expression of myocardial nuclear NF-κB-p65 and caspase-3 which were markedly inhibited by trifluoperazine, suggesting that cardioprotection conferred by trifluoperazine involved, at least in part, suppression of NF-κB and apoptosis. Furthermore, biochemical and histopathological examinations showed that trifluoperazine improved doxorubicin-induced renal and hepatic impairments both functionally and structurally. SIGNIFICANCE In conclusion, the present in vivo study is the first to provide evidences underscoring the protective effects of trifluoperazine that may pave the way for repurposing this calmodulin antagonist in ameliorating organ toxicity by doxorubicin.
Collapse
Affiliation(s)
- Ahmed E Goda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt.
| | - Amr M Elenany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt
| | - Alaa E Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt
| |
Collapse
|
14
|
Vanhoutte D, Schips TG, Vo A, Grimes KM, Baldwin TA, Brody MJ, Accornero F, Sargent MA, Molkentin JD. Thbs1 induces lethal cardiac atrophy through PERK-ATF4 regulated autophagy. Nat Commun 2021; 12:3928. [PMID: 34168130 PMCID: PMC8225674 DOI: 10.1038/s41467-021-24215-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/31/2021] [Indexed: 02/05/2023] Open
Abstract
The thrombospondin (Thbs) family of secreted matricellular proteins are stress- and injury-induced mediators of cellular attachment dynamics and extracellular matrix protein production. Here we show that Thbs1, but not Thbs2, Thbs3 or Thbs4, induces lethal cardiac atrophy when overexpressed. Mechanistically, Thbs1 binds and activates the endoplasmic reticulum stress effector PERK, inducing its downstream transcription factor ATF4 and causing lethal autophagy-mediated cardiac atrophy. Antithetically, Thbs1-/- mice develop greater cardiac hypertrophy with pressure overload stimulation and show reduced fasting-induced atrophy. Deletion of Thbs1 effectors/receptors, including ATF6α, CD36 or CD47 does not diminish Thbs1-dependent cardiac atrophy. However, deletion of the gene encoding PERK in Thbs1 transgenic mice blunts the induction of ATF4 and autophagy, and largely corrects the lethal cardiac atrophy. Finally, overexpression of PERK or ATF4 using AAV9 gene-transfer similarly promotes cardiac atrophy and lethality. Hence, we identified Thbs1-mediated PERK-eIF2α-ATF4-induced autophagy as a critical regulator of cardiomyocyte size in the stressed heart.
Collapse
Affiliation(s)
- Davy Vanhoutte
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tobias G Schips
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Janssen Pharmaceuticals, Spring House, PA, USA
| | - Alexander Vo
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kelly M Grimes
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tanya A Baldwin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew J Brody
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Federica Accornero
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Michelle A Sargent
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
15
|
Nicol M, Sadoune M, Polidano E, Launay JM, Samuel JL, Azibani F, Cohen-Solal A. Doxorubicin-induced and trastuzumab-induced cardiotoxicity in mice is not prevented by metoprolol. ESC Heart Fail 2021; 8:928-937. [PMID: 33529501 PMCID: PMC8006653 DOI: 10.1002/ehf2.13198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 12/04/2020] [Accepted: 12/28/2020] [Indexed: 01/01/2023] Open
Abstract
Aims Our objectives were to validate a murine model of chronic cardiotoxicity induced by Doxorubicin (Dox) and Trastuzumab (Trast) and to test the potential cardio‐protective effect of metoprolol. Methods and results Male C57Bl6 mice were intraperitoneally injected during 2 weeks with Dox (24 mg/kg) or saline, and then with Trast (10 mg/kg) or saline for two more weeks. Half of the mice received metoprolol (100 mg/kg). Cardiotoxicity was defined by a decline in left ventricular ejection fraction (LVEF) ≥ 10 points. At Day 42, Dox + Trast‐treated mice exhibited a 13‐points decline in LVEF (74 ± 2.6% vs. 87 ± 0.8% for control mice, P < 0.001) and a severe cardiac atrophy (heart weight: 105 ± 2.7 mg vs. 119 ± 3.9 mg for control mice, P < 0.01). This cardiac atrophy resulted from an excess of cardiac necrosis (assessed by plasma cardiac troponin I level: 3.2 ± 0.4 ng/L vs. 1.3 ± 0.06 ng/L for control mice, P < 0.01), an increase in apoptosis (caspase 3 activity showing a six‐fold increase for Dox + Trast‐treated mice vs. controls, P < 0.001), and cardiomyocyte atrophy (myocyte size: 0.67 ± 0.08 μm2 vs. 1.36 ± 0.10 μm2 for control mice, P < 0.001). In addition, Dox + Trast‐treated mice were shown to have an increased cardiac oxidative stress (164 ± 14 dihydroethidine‐marked nuclei per area vs. 56 ± 9.5 for control mice, P < 0.01) and increased cardiac fibrosis (the semi‐quantitative fibrosis score was three‐fold higher for Dox + Trast‐treated mice as compared with controls, P < 0.01). Metoprolol was not able to prevent either the decrease in LVEF or the severe cardiac atrophy, the cardiac necrosis, and the cardiac remodelling induced by chemotherapies. Conclusion A murine model of chronic cardiotoxicity induced by Dox and Trast was characterized by a decrease in cardiac function, a cardiac apoptosis and necrosis leading to cardiomyocyte atrophy. Metoprolol did not prevent this cardiotoxicity.
Collapse
Affiliation(s)
- Martin Nicol
- Cardiology Department, Lariboisiere Hospital, University of Paris, Paris, France.,Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Malha Sadoune
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Evelyne Polidano
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Jean Marie Launay
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Jane Lise Samuel
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Feriel Azibani
- Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| | - Alain Cohen-Solal
- Cardiology Department, Lariboisiere Hospital, University of Paris, Paris, France.,Inserm UMR-S 942, University of Paris, Lariboisiere Hospital, Paris, France
| |
Collapse
|
16
|
Baumfalk DR, Opoku-Acheampong AB, Caldwell JT, Butenas ALE, Horn AG, Kunkel ON, Copp SW, Ade CJ, Musch TI, Behnke BJ. Effects of high-intensity training on prostate cancer-induced cardiac atrophy. Am J Transl Res 2021; 13:197-209. [PMID: 33527018 PMCID: PMC7847523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Recent evidence suggests prostate cancer independent of treatment has atrophic effects on whole heart and left ventricular (LV) masses, associated with reduced endurance exercise capacity. In a pre-clinical model, we tested the hypothesis that high-intensity training could prevent cardiac atrophy with prostate cancer and alter cardiac protein degradation mechanisms. METHODS Dunning R-3327 AT-1 prostate cancer cells (1×105) were injected into the ventral prostate lobe of 5-6 mo immunocompetent Copenhagen rats (n=24). These animals were randomized into two groups, tumor-bearing exercise (TBEX, n=15) or tumor bearing sedentary (TBS, n=9). Five days after surgery, TBEX animals began exercise on a treadmill (25 m/min, 15° incline) for 45-60 min/day for 18±2 days. Pre-surgery (Pre), and post-exercise training (Post) echocardiographic evaluation (Vivid S6, GE Health Care), using the parasternal short axis view, was used to examine ventricle dimensions. Markers of protein degradation (muscle atrophy F-box, Cathepsin B, Cathepsin L) in the left ventricle were semi-quantified via Western Blot. RESULTS There were no significant differences in tumor mass between groups (TBEX 3.4±0.7, TBS 2.8±0.6 g, P=0.3), or body mass (TBEX 317±5, TBS 333±7 g, P=0.2). Heart-to-body mass ratio was lower in TBS group compared to TBEX (2.3±0.1 vs. 2.5±0.1 mg/g, P<0.05). LV/body mass ratio was also lower in the TBS group (1.6±0.1 vs. 1.8±0.1 mg/g, P<0.05). From Pre-Post, TBEX had significant increases in SV (~20% P<0.05) whereas TBS had no significant change. There were no significant differences between groups for markers of protein degradation. CONCLUSION This study suggests that high-intensity exercise can improve LV function and increase LV mass concurrent with prostate cancer development, versus sedentary counterparts. Given cardiac dysfunction often manifests with conventional anti-cancer treatments, a short-term high-intensity training program, prior to treatment, may improve cardiac function and fatigue resistance in cancer patients.
Collapse
Affiliation(s)
- Dryden R Baumfalk
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
| | | | - Jacob T Caldwell
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
| | - Alec L E Butenas
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
| | - Andrew G Horn
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
| | - Olivia N Kunkel
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
| | - Steven W Copp
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
| | - Carl J Ade
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
- Johnson Cancer Research Center, Kansas State UniversityManhattan 66506, Kansas, USA
| | - Timothy I Musch
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
- Department of Anatomy and Physiology, Kansas State UniversityManhattan 66506, Kansas, USA
| | - Bradley J Behnke
- Department of Kinesiology, Kansas State UniversityManhattan 66506, Kansas, USA
- Johnson Cancer Research Center, Kansas State UniversityManhattan 66506, Kansas, USA
| |
Collapse
|
17
|
Miyagawa Y, Nukaga S, Mori T, Fujiwara-Tani R, Fujii K, Mori S, Goto K, Kishi S, Sasaki T, Nakashima C, Ohmori H, Kawahara I, Luo Y, Kuniyasu H. Evaluation of cancer-derived myocardial impairments using a mouse model. Oncotarget 2020; 11:3712-3722. [PMID: 33110478 PMCID: PMC7566807 DOI: 10.18632/oncotarget.27759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/10/2020] [Indexed: 01/21/2023] Open
Abstract
Myocardial damage in cancer patients is emphasized as a cause of death; however, there are not many murine cachexia models to evaluate cancer-derived heart disorder. Using the mouse cachexia model that we established previously, we investigated myocardial damage in tumor-bearing mice. In cachexic mice, decreased heart weight and myocardial volume, and dilated left ventricular lumen, and atrophied cardiomyocytes were noted. The cardiomyocytes also showed accumulated 8-hydroxydeoxyguanosine, decreased leucine zipper and EF-hand-containing transmembrane protein-1, and increased microtubule-associated protein light chain3-II. Levels of tumor necrosis factor-α and high-mobility group box-1 proteins in the myocardium were increased, and nuclear factor κB, a signaling molecule associated with these proteins, was activated. When rat cardiomyoblasts (H9c2 cells) were treated with mouse cachexia model ascites and subjected to flux analysis, both oxidative phosphorylation and glycolysis were suppressed, and the cells were in a quiescent state. These results are in good agreement with those previously reported on cancerous myocardial damage. The established mouse cachexia model can therefore be considered useful for analyzing cancer-derived myocardial damage.
Collapse
Affiliation(s)
- Yoshihiro Miyagawa
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Shota Nukaga
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan.,Division of Rehabilitation, Hanna Central Hospital, Ikoma, Nara 630-0243, Japan
| | - Takuya Mori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kei Goto
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan.,Division of Rehabilitation, Hoshida Minami Hospital, Katano, Osaka 576-0022, Japan
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Chie Nakashima
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Isao Kawahara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan.,Division of Rehabilitation, Hanna Central Hospital, Ikoma, Nara 630-0243, Japan
| | - Yi Luo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| |
Collapse
|
18
|
Bracun V, Aboumsallem JP, van der Meer P, de Boer RA. Cardiac Biomarkers in Patients with Cancer: Considerations, Clinical Implications, and Future Avenues. Curr Oncol Rep 2020; 22:67. [PMID: 32514994 PMCID: PMC7280346 DOI: 10.1007/s11912-020-00930-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF THE REVIEW As the number of cancer survivors increases due to early screening and modern (antineoplastic) treatments, cancer treatment associated cardiotoxicity (CTAC) is becoming an increasing health burden that affects survival and quality of life among cancer survivors. Thus, clinicians need to identify adverse events early, in an effort to take suitable measures before the occurrence of permanent or irreversible cardiac dysfunction. RECENT FINDINGS Cardiac troponin (cTn) and B-type natriuretic peptide (BNP) have been proven to detect subclinical cardiotoxicity during antineoplastic treatment. As such, these cardio-specific biomarkers could predict which patients are at risk of developing CTAC even before the start of therapy. Nevertheless, there are inconsistent data from published studies, and the recommendations regarding the use of these biomarkers and their validity are mostly based on expert consensus opinion. In this review, we summarize available literature that evaluates biomarkers of CTAC, and we encourage strategies that integrate circulating biomarkers and cardiac imaging in identifying cancer patients that are at high risk.
Collapse
Affiliation(s)
- Valentina Bracun
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, PO Box 30.001, 9700 RB Groningen, the Netherlands
| |
Collapse
|
19
|
Oishi Y, Manabe I. Organ System Crosstalk in Cardiometabolic Disease in the Age of Multimorbidity. Front Cardiovasc Med 2020; 7:64. [PMID: 32411724 PMCID: PMC7198858 DOI: 10.3389/fcvm.2020.00064] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
The close association among cardiovascular, metabolic, and kidney diseases suggests a common pathological basis and significant interaction among these diseases. Metabolic syndrome and cardiorenal syndrome are two examples that exemplify the interlinked development of disease or dysfunction in two or more organs. Recent studies have been sorting out the mechanisms responsible for the crosstalk among the organs comprising the cardiovascular, metabolic, and renal systems, including heart-kidney and adipose-liver signaling, among many others. However, it is also becoming clear that this crosstalk is not limited to just pairs of organs, and in addition to organ-organ crosstalk, there are also organ-system and organ-body interactions. For instance, heart failure broadly impacts various organs and systems, including the kidney, liver, lung, and nervous system. Conversely, systemic dysregulation of metabolism, immunity, and nervous system activity greatly affects heart failure development and prognosis. This is particularly noteworthy, as more and more patients present with two or more coexisting chronic diseases or conditions (multimorbidity) due in part to the aging of society. Advances in treatment also contribute to the increase in multimorbidity, as exemplified by cardiovascular disease in cancer survivors. To understand the mechanisms underlying the increasing burden of multimorbidity, it is vital to elucidate the multilevel crosstalk and communication within the body at the levels of organ systems, tissues, and cells. In this article, we focus on chronic inflammation as a key common pathological basis of cardiovascular and metabolic diseases, and discuss emerging mechanisms that drive chronic inflammation in the context of multimorbidity.
Collapse
Affiliation(s)
- Yumiko Oishi
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
20
|
Li M, Russo M, Pirozzi F, Tocchetti CG, Ghigo A. Autophagy and cancer therapy cardiotoxicity: From molecular mechanisms to therapeutic opportunities. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118493. [DOI: 10.1016/j.bbamcr.2019.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/31/2019] [Accepted: 06/15/2019] [Indexed: 11/25/2022]
|
21
|
Obrezan AG, Shcherbakova NV. [Pathogenetic mechanisms of development of myocardial pathology in patients with malignant tumors: the current state of the problem]. ACTA ACUST UNITED AC 2020; 60:142-154. [PMID: 32345210 DOI: 10.18087/cardio.2020.2.n985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 12/23/2019] [Indexed: 11/18/2022]
Abstract
The myocardium, which has a high metabolic activity, responds to metabolic disorders and energy imbalance induced by a growing malignant tumor. In addition, the tumor itself can produce substances that directly affect metabolic processes and the life cycle of cells not involved in the neoplastic process, including cardiomyocytes. This review summarized and systematized current data on individual aspects of detrimental effects of oncogenes and tumor-related factors on the heart muscle and morpho-functional changes in the cardiovascular system of oncology patients. Also, the authors described in detail development of these pathogenetic mechanisms.
Collapse
Affiliation(s)
- A G Obrezan
- St. Petersburg State University, SOGAZ International Medical Center, St. Petersburg
| | | |
Collapse
|
22
|
Abstract
Cachexia is a multifactorial disease characterized by a pathologic shift of metabolism towards a more catabolic state. It frequently occurs in patients with chronic diseases such as chronic heart failure and is especially common in the elderly. In patients at risk, cardiac cachexia is found in about 10% of heart failure patients. The negative impact of cardiac cachexia on mortality, morbidity, and quality of life demonstrates the urgent need to find new effective therapies against cardiac cachexia. Furthermore, exercise training and nutritional support can help patients with cardiac cachexia. Despite ongoing efforts to find new therapies for cachexia treatment, also new preventive strategies are needed.
Collapse
Affiliation(s)
- Alessia Lena
- Division of Cardiology and Metabolism, Department of Cardiology, Charité-Campus Virchow Klinikum (CVK), Augustenburger Platz 1, 13353 Berlin, Germany.,Department of Cardiology, Charité-Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Föhrer Str. 15, 13353 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
| | - Nicole Ebner
- Department of Cardiology, University Medical Center Goettingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Markus S Anker
- Division of Cardiology and Metabolism, Department of Cardiology, Charité-Campus Virchow Klinikum (CVK), Augustenburger Platz 1, 13353 Berlin, Germany.,Department of Cardiology, Charité-Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Föhrer Str. 15, 13353 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
| |
Collapse
|
23
|
Zhang Y, Knight W, Chen S, Mohan A, Yan C. Multiprotein Complex With TRPC (Transient Receptor Potential-Canonical) Channel, PDE1C (Phosphodiesterase 1C), and A2R (Adenosine A2 Receptor) Plays a Critical Role in Regulating Cardiomyocyte cAMP and Survival. Circulation 2019; 138:1988-2002. [PMID: 29871977 DOI: 10.1161/circulationaha.118.034189] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND cAMP plays a critical role in regulating cardiomyocyte survival. Various cAMP signaling pathways behave distinctly or in opposition. We have previously reported that activation of cAMP hydrolysis by cyclic nucleotide phosphodiesterase 1C (PDE1C) promotes cardiomyocytes death/apoptosis, yet the underlying molecular mechanism remains unknown. In this study, we aimed to identify the specific cAMP signaling pathway modulated by PDE1C and determine the mechanism by which Ca2+/calmodulin-stimulated PDE1C is activated. METHODS To study cardiomyocyte death/apoptosis, we used both isolated mouse adult cardiomyocytes in vitro and doxorubicin-induced cardiotoxicity in vivo. We used a variety of pharmacological activators and inhibitors as well as genetically engineered molecular tools to manipulate the expression and activity of proteins of interest. RESULTS We found that the protective effect of PDE1C inhibition/deficiency on Ang II or doxorubicin-induced cardiomyocyte death/apoptosis is dependent on cAMP-generating adenosine A2 receptors (A2Rs), suggesting that PDE1C's cAMP-hydrolyzing activity selectively modulates A2R-cAMP signaling in cardiomyocytes. In addition, we found that the effects of PDE1C activation on Ang II-mediated cAMP reduction and cardiomyocyte death are dependent on transient receptor potential-canonical (TRPC) channels, in particular TRPC3. We also observed synergistic protective effects on cardiomyocyte survival from the combination of A2R stimulation together with PDE1 or TRPC inhibition. Coimmunostaining and coimmunoprecipitation studies showed that PDE1C is localized in proximity with A2R and TRPC3 in the plasma membrane and perhaps T tubules. It is important to note that we found that doxorubicin-induced cardiac toxicity and dysfunction in mice are attenuated by the PDE1 inhibitor IC86340 or in PDE1C knockout mice, and this protective effect is significantly diminished by A2R antagonism. CONCLUSIONS We have characterized a novel multiprotein complex comprised of A2R, PDE1C, and TRPC3, in which PDE1C is activated by TRPC3-derived Ca2+, thereby antagonizing A2R-cAMP signaling and promoting cardiomyocyte death/apoptosis. Targeting these molecules individually or in combination may represent a compelling therapeutic strategy for potentiating cardiomyocyte survival.
Collapse
Affiliation(s)
- Yishuai Zhang
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Walter Knight
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY.,Department of Pharmacology and Physiology (W.K., S.C.), University of Rochester School of Medicine and Dentistry, NY
| | - Si Chen
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY.,Department of Pharmacology and Physiology (W.K., S.C.), University of Rochester School of Medicine and Dentistry, NY
| | - Amy Mohan
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| |
Collapse
|
24
|
Pietzsch S, Ricke-Hoch M, Stapel B, Hilfiker-Kleiner D. Modulation of cardiac AKT and STAT3 signalling in preclinical cancer models and their impact on the heart. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118519. [PMID: 31374232 DOI: 10.1016/j.bbamcr.2019.07.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Advanced cancer induces fundamental cardiac changes and promotes body wasting and heart failure. We evaluated the impact of cancer on major cardiac signalling pathways, and resulting consequences for the heart. METHODS AND RESULTS Metastatic melanoma disease was induced in male C57BL/6 N mice by intraperitoneal injection of the melanoma cell line B16F10 and lead to cardiac atrophy and heart failure. Analyses of key cardiac signalling pathways in left ventricular tissue revealed increased activation of STAT3 and reduced activation of AKT, p38 and ERK1/2. Markers of the ubiquitin proteasomal system (UPS: Atrogin-1) and of mitophagy/autophagy (LC3b, BNIP3) were upregulated. Tumour-bearing C57BL/6 N mice with a cardiomyocyte-specific overexpression of a constitutively active AKT transgene (AKTtg) displayed less cardiac atrophy and dysfunction and normalized Atrogin-1, LC3b and BNIP3 expression while the cardiomyocyte-specific knockout of STAT3 (CKO) had no major effect on these parameters compared to WT. CONCLUSION Cancer alters major cardiac signalling pathways and subsequently the UPS, mitophagy and autophagy. The present study suggests that cancer-induced reduction of cardiomyocyte AKT contributes to these alterations as they were attenuated in tumour-bearing AKTtg mice. In turn, increased cardiomyocyte STAT3 activation appears less relevant, as tumour-induced impairment on the heart was largely similar in CKO and WT mice. Since oncologic therapies frequently target AKT and/or STAT3, their impact on the heart might be different in tumour-bearing mice compared to healthy mice, a feature suggesting to test tumour therapies also in tumour disease models and not only under healthy conditions. This article is part of a Special Issue entitled: Cardiomyocyte biology: new pathways of differentiation and regeneration edited by Marijke Brink, Marcus C. Schaub, and Christian Zuppinger.
Collapse
Affiliation(s)
- Stefan Pietzsch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Britta Stapel
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
25
|
Levick SP, Soto-Pantoja DR, Bi J, Hundley WG, Widiapradja A, Manteufel EJ, Bradshaw TW, Meléndez GC. Doxorubicin-Induced Myocardial Fibrosis Involves the Neurokinin-1 Receptor and Direct Effects on Cardiac Fibroblasts. Heart Lung Circ 2018; 28:1598-1605. [PMID: 30205930 DOI: 10.1016/j.hlc.2018.08.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/02/2018] [Accepted: 08/14/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cancer patients receiving anthracycline-based chemotherapy (Anth-bC) may experience early cardiac fibrosis, which could be an important contributing mechanism to the development of impaired left ventricular (LV) function. Substance P, a neuropeptide that predominantly acts via the neurokinin 1 receptor (NK-1R), contributes to adverse myocardial remodelling and fibrosis in other cardiomyopathies. We sought to determine if NK-1R blockade is effective against doxorubicin (Dox - a frequently used Anth-bC)-induced cardiac fibrosis and cardiomyocyte apoptosis. In addition, we explored the direct effects of Dox on cardiac fibroblasts. METHODS Male Sprague-Dawley rats were randomised to receive saline, six cycles of Dox (1.5mg Dox/kg/cycle) or Dox with an NK-1R antagonist (L732138, 5mg/kg/daily through Dox treatment). At 8 weeks after the initial dose of Dox, LV function and histopathological myocardial fibrosis and cell apoptosis were assessed. Collagen secretion was measured in vitro to test direct Dox activation of cardiac fibroblasts. RESULTS Rats undergoing Dox treatment (9mg/kg cumulative dose) developed cardiac fibrosis and cardiomyocyte apoptosis. NK-1R blockade partially mitigated cardiac fibrosis while completely preventing cardiomyocyte apoptosis. This resulted in improved diastolic function. Furthermore, we found that Dox had direct effects on cardiac fibroblasts to cause increased collagen production and enhanced cell survival. CONCLUSIONS This study demonstrates that cardiac fibrosis induced by Anth-bC can be reduced by NK-1R blockade. The residual fibrotic response is likely due to direct Dox effects on cardiac fibroblasts to produce collagen.
Collapse
Affiliation(s)
- Scott P Levick
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David R Soto-Pantoja
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA; Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jianli Bi
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - W Gregory Hundley
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Radiological Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Alexander Widiapradja
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Edward J Manteufel
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tancia W Bradshaw
- Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Giselle C Meléndez
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
26
|
Antunes J, Ferreira RM, Moreira-Gonçalves D. Exercise Training as Therapy for Cancer-Induced Cardiac Cachexia. Trends Mol Med 2018; 24:709-727. [DOI: 10.1016/j.molmed.2018.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 12/27/2022]
|