1
|
Kyriakopoulou E, van Kampen SJ, Wehrens M, Han SJ, de Ruiter H, Monshouwer-Kloots J, Marshall E, Brodehl A, van der Kraak P, te Riele AS, van Aarnhem EE, van Laake LW, Tsui H, Boogerd CJ, van Rooij E. EPAS1 induction drives myocardial degeneration in desmoplakin-cardiomyopathy. iScience 2025; 28:111895. [PMID: 40034852 PMCID: PMC11872638 DOI: 10.1016/j.isci.2025.111895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/24/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is frequently attributed to desmosomal mutations, such as those in the desmoplakin (DSP) gene. Patients with DSP-cardiomyopathy are predisposed to myocardial degeneration and arrhythmias. Despite advancements, the underlying molecular mechanisms remain incompletely understood, thus limiting therapeutic options. Here, we employed spatial transcriptomics on an explanted heart from a patient with a pathogenic DSP variant. Our transcriptional analysis revealed endothelial PAS domain-containing protein 1 (EPAS1) as a potential regulator of mitochondrial homeostasis in stressed cardiomyocytes. Elevated EPAS1 levels were associated with mitochondrial dysfunction and hypoxic stress in both human-relevant in vitro ACM models and additional explanted hearts with genetic cardiomyopathy. Collectively, cardiomyocytes bearing pathogenic DSP variants exhibit mitochondrial dysfunction, increased apoptosis, and impaired contractility, which are linked to the increased EPAS1 levels. These findings implicate EPAS1 as a key regulator of myocardial degeneration in DSP-cardiomyopathy, which expand to other forms of ACM.
Collapse
Affiliation(s)
- Eirini Kyriakopoulou
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sebastiaan J. van Kampen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Martijn Wehrens
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Su Ji Han
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hesther de Ruiter
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Emma Marshall
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Andreas Brodehl
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Petra van der Kraak
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anneline S.J.M. te Riele
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Egidius E.H.L. van Aarnhem
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Linda W. van Laake
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hoyee Tsui
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cornelis J. Boogerd
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
2
|
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
|
3
|
Ciccarese C, Anghelone A, Stefani A, Cigliola A, Strusi A, D'Agostino F, Bria E, Iacovelli R, Tortora G. The incidence and relative risk of major adverse cardiovascular events and hypertension in patients treated with immune checkpoint inhibitors plus tyrosine-kinase inhibitors for solid tumors: a systemic review and meta-analysis. Expert Rev Anticancer Ther 2024; 24:623-633. [PMID: 38879826 DOI: 10.1080/14737140.2024.2357814] [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: 03/08/2023] [Accepted: 05/03/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION Combinations of immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) can be responsible for major adverse cardiovascular events (MACEs). We performed a meta-analysis to assess the relative risk (RR) of MACEs and hypertension in cancer patients treated with ICI+TKI combinations. RESEARCH DESIGN AND METHODS We selected prospective trials through MEDLINE/PubMed, Cochrane Library, and ASCOMeeting abstracts. We calculated combined ORs, RRs, and 95% CIs using RevMansoftware for meta-analysis (v.5.2.3). RESULTS Seven studies were selected for the analysis of MACEs (3849 patients). The incidence MACEs were 0.8% with ICI+TKI combinations, compared to 0.2% in the control arms for both any- and high-grade. ICI+TKI combinations significantly increased the risk of any- (OR = 3.21; p = 0.01) and high-grade MACEs (OR = 2.72; p = 0.05). Ten studies were selected for the analysis of hypertension (5744 patients). The incidence of treatment-related hypertension of any-grade and high-grade was41.3% (vs. 20.8%) and 26.1% (vs. 12.3%) with ICI+TKI combinations, respectively. ICI+TKI combinations significantly increased the risk of treatment-related hypertension of any-grade (RR = 2.10; p = 0.001), but not of high-grade (p = 0.11). CONCLUSIONS ICI+TKI combinations increase the risk of MACEs compared to controls, although the absolute incidence is eventually low. Routine cardiovascular monitoring in asymptomatic patients is therefore not recommended.
Collapse
Affiliation(s)
- Chiara Ciccarese
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Alessio Stefani
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Cigliola
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Emilio Bria
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Iacovelli
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
4
|
Kreidieh F, McQuade J. Novel insights into cardiovascular toxicity of cancer targeted and immune therapies: Beyond ischemia with non-obstructive coronary arteries (INOCA). AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 40:100374. [PMID: 38510501 PMCID: PMC10946000 DOI: 10.1016/j.ahjo.2024.100374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Novel immune and targeted therapies approved over the past 2 decades have resulted in dramatic improvements in cancer-specific outcomes for many cancer patients. However, many of these agents can induce cardiovascular toxicity in a subset of patients. The field of cardio-oncology was established based on observations that anti-neoplastic chemotherapies and mantle radiation can lead to premature cardiomyopathy in cancer survivors. While conventional chemotherapy, targeted therapy, and immune therapies can all result in cardiovascular adverse events, the mechanisms, timing, and incidence of these events are inherently different. Many of these effects converge upon the coronary microvasculature to involve, through endocardial endothelial cells, a more direct effect through close proximity to cardiomyocyte with cellular communication and signaling pathways. In this review, we will provide an overview of emerging paradigms in the field of Cardio-Oncology, particularly the role of the coronary microvasculature in mediating cardiovascular toxicity of important cancer targeted and immune therapies. As the number of cancer patients treated with novel immune and targeted therapies grows exponentially and subsequently the number of long-term cancer survivors dramatically increases, it is critical that cardiologists and cardiology researchers recognize the unique potential cardiovascular toxicities of these agents.
Collapse
Affiliation(s)
- Firas Kreidieh
- Instructor of Clinical Medicine- Division of Hematology-Oncology; Associate Director- Internal Medicine Residency Program, American University of Beirut, Beirut, Lebanon
| | - Jennifer McQuade
- Associate Professor and Physician Scientist in Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
5
|
Di Marco A, Artioli G, Favaretto A, Cavasin N, Basso U. Multiorgan failure caused by pembrolizumab and axitinib in a woman affected by metastatic clear cell renal cell carcinoma: A case report and literature review. Medicine (Baltimore) 2024; 103:e37606. [PMID: 38552059 PMCID: PMC10977559 DOI: 10.1097/md.0000000000037606] [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: 12/12/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024] Open
Abstract
RATIONALE Treatment with a combination of immune checkpoint inhibitors (ICIs) (pembrolizumab or nivolumab) and oral Tyrosine Kinase Inhibitors (TKI) targeting angiogenesis (axitinib, cabozantinib or lenvatinib) has shown benefits in terms of efficacy and survival in metastatic renal cell carcinoma (mRCC), with a favorable toxicity profile. However, some rare and serious treatment-related adverse events can be difficult to manage. PATIENT CONCERNS Here we report the first case of an mRCC patient who, after only 2 administrations of pembrolizumab-axitinib, experienced severe multiorgan failure (MOF) with heart failure, oliguria and acute hepatitis requiring aggressive supportive treatment in intensive care unit. DIAGNOSES A diagnosis of severe MOF induced by pembrolizumab plus axitinib was considered. INTERVENTIONS The patient was treated with dobutamine, levosimendan along with high-dose steroids under continuous cardiologic monitoring. OUTCOMES After treatment, the patient had a full recovery and was discharged from the hospital. LESSONS We reviewed all the other cases of MOF reported during treatment with combined ICI-TKI in cancer patients in order to summarize incidence, clinical manifestations and management with a specific focus on the need for prompt recognition and aggressive management under multidisciplinary care.
Collapse
Affiliation(s)
- Andrea Di Marco
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Grazia Artioli
- Department of Medical Oncology, AULSS 2 Marca Trevigiana, Ca’ Foncello Hospital, Treviso, Italy
| | - Adolfo Favaretto
- Department of Medical Oncology, AULSS 2 Marca Trevigiana, Ca’ Foncello Hospital, Treviso, Italy
| | - Nicolò Cavasin
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Umberto Basso
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padua, Italy
| |
Collapse
|
6
|
Krumbein M, Oberman F, Cinnamon Y, Golomb M, May D, Vainer G, Belzer V, Meir K, Fridman I, Haybaeck J, Poelzl G, Kehat I, Beeri R, Kessler SM, Yisraeli JK. RNA binding protein IGF2BP2 expression is induced by stress in the heart and mediates dilated cardiomyopathy. Commun Biol 2023; 6:1229. [PMID: 38052926 PMCID: PMC10698010 DOI: 10.1038/s42003-023-05547-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] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
The IGF2BP family of RNA binding proteins consists of three paralogs that regulate intracellular RNA localization, RNA stability, and translational control. Although IGF2BP1 and 3 are oncofetal proteins, IGF2BP2 expression is maintained in many tissues, including the heart, into adulthood. IGF2BP2 is upregulated in cardiomyocytes during cardiac stress and remodeling and returns to normal levels in recovering hearts. We wondered whether IGF2BP2 might play an adaptive role during cardiac stress and recovery. Enhanced expression of an IGF2BP2 transgene in a conditional, inducible mouse line leads to dilated cardiomyopathy (DCM) and death within 3-4 weeks in newborn or adult hearts. Downregulation of the transgene after 2 weeks, however, rescues these mice, with complete recovery by 12 weeks. Hearts overexpressing IGF2BP2 downregulate sarcomeric and mitochondrial proteins and have fragmented mitochondria and elongated, thinner sarcomeres. IGF2BP2 is also upregulated in DCM or myocardial infarction patients. These results suggest that IGF2BP2 may be an attractive target for therapeutic intervention in cardiomyopathies.
Collapse
Affiliation(s)
- Miriam Krumbein
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Froma Oberman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Cinnamon
- Institute of Animal Science, Agricultural Research Organization, The Volcani Institute, Rishon Lezion, Israel
| | | | - Dalit May
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Shaare Zedek Medical Center, Jerusalem, Israel
- Clalit Health Service, Jerusalem, Israel
| | - Gilad Vainer
- Department of Pathology, Hadassah Medical Center, Jerusalem, Israel
| | - Vitali Belzer
- Department of Pathology, Hadassah Medical Center, Jerusalem, Israel
| | - Karen Meir
- Department of Pathology, Hadassah Medical Center, Jerusalem, Israel
| | - Irina Fridman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Johannes Haybaeck
- Institut für Pathologie, Neuropathologie und Molekularpathologie, Medical University Innsbruck, Innsbruck, Austria
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8010, Graz, Austria
| | - Gerhard Poelzl
- Department of Cardiology and Angiology, Medical University Innsbruck, Innsbruck, Austria
| | - Izhak Kehat
- Department of Physiology and Biophysics, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Bat Galim, Haifa, Israel
| | - Ronen Beeri
- Department of Cardiology, Hadassah Medical Center, Jerusalem, Israel
| | - Sonja M Kessler
- Experimental Pharmacology for Natural Sciences, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | - Joel K Yisraeli
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
7
|
Sager R, Keller LS, Stehli J, Jakob P, Michel J, Kasel M, Templin C, Stähli BE. Association of N-terminal pro-B-type natriuretic peptide with mortality in elderly (≥80 years) patients undergoing percutaneous coronary intervention. Catheter Cardiovasc Interv 2022; 100:535-543. [PMID: 36040724 DOI: 10.1002/ccd.30365] [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: 03/06/2022] [Revised: 07/06/2022] [Accepted: 07/27/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVES The aim of this study was to determine the role of N-terminal pro-B-type natriuretic peptide (NT-proBNP) in the prognostication of patients ≥80 years of age undergoing percutaneous coronary intervention (PCI). BACKGROUND Elderly patients with coronary artery disease in need of PCI represent a growing patient population. Advanced risk prediction in this frail and comorbid patient population is important. METHODS A total of 460 consecutive patients ≥80 years of age undergoing PCI for acute (ACS) or chronic coronary syndromes (CCS) at the University Hospital Zurich, Switzerland, between January 2016 and December 2018 and with available baseline NT-proBNP levels were included in the analysis. Patients were stratified according to baseline NT-proBNP levels. The primary endpoint was all-cause mortality at a median follow-up of 33 (interquartile range: 3-392) days. RESULTS Median baseline NT-proBNP levels were 1411 (457-3984) ng/L. All-cause mortality was 7.8% in the lowest and 27.8% in the highest NT-proBNP quartile group (p < 0.001). In patients with ACS, all-cause mortality was 4.8% and 30.4% in the lowest and the highest NT-proBNP quartile (p < 0.001), and corresponding rates in patients with CCS were 11.1% and 22.2% (p = 0.38). In multivariable Cox regression analysis, baseline NT-proBNP levels were independently associated with an increased risk of all-cause mortality (adjusted hazard ratio: 1.00, 95% confidence interval: 1.00-1.00, p = 0.04). CONCLUSIONS Baseline NT-proBNP levels were identified as independent predictor of mortality in elderly (≥80 years) patients undergoing PCI. Hence, baseline NT-proBNP allows for the identification of a high-risk elderly patient subset.
Collapse
Affiliation(s)
- Raphael Sager
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Lukas S Keller
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Julia Stehli
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Philipp Jakob
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Jonathan Michel
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Markus Kasel
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Christian Templin
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| | - Barbara E Stähli
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Cardio-onco-metabolism: metabolic remodelling in cardiovascular disease and cancer. Nat Rev Cardiol 2022; 19:414-425. [PMID: 35440740 PMCID: PMC10112835 DOI: 10.1038/s41569-022-00698-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease and cancer are the two leading causes of morbidity and mortality in the world. The emerging field of cardio-oncology has revealed that these seemingly disparate disease processes are intertwined, owing to the cardiovascular sequelae of anticancer therapies, shared risk factors that predispose individuals to both cardiovascular disease and cancer, as well the possible potentiation of cancer growth by cardiac dysfunction. As a result, interest has increased in understanding the fundamental biological mechanisms that are central to the relationship between cardiovascular disease and cancer. Metabolism, appropriate regulation of energy, energy substrate utilization, and macromolecular synthesis and breakdown are fundamental processes for cellular and organismal survival. In this Review, we explore the emerging data identifying metabolic dysregulation as an important theme in cardio-oncology. We discuss the growing recognition of metabolic reprogramming in cardiovascular disease and cancer and view the novel area of cardio-oncology through the lens of metabolism.
Collapse
|
9
|
Chowdhury K, Lai SL, Marín-Juez R. Modulation of VEGFA Signaling During Heart Regeneration in Zebrafish. Methods Mol Biol 2022; 2475:297-312. [PMID: 35451767 DOI: 10.1007/978-1-0716-2217-9_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the last decades, myocardial infarction and heart failure have accounted every year for millions of deaths worldwide. After a coronary occlusion, the lack of blood supply to downstream muscle leads to cell death and scarring. To date, several pro-angiogenic factors have been tested to stimulate reperfusion of the affected myocardium, VEGFA being one of the most extensively studied. Given the unsuccessful outcomes of clinical trials, understanding how cardiac revascularization takes place in models with endogenous regenerative capacity holds the key to devising more efficient therapies. Here, we summarize the main findings on VEGFA's role during cardiac repair and regeneration, with a particular focus on zebrafish as a regenerative model. Moreover, we provide a comprehensive overview of available tools to modulate Vegfa expression and action in zebrafish regeneration studies. Understanding the role of Vegfa during zebrafish heart regeneration may help devise efficient therapies and circumvent current limitations in using VEGFA for therapeutic angiogenesis approaches.
Collapse
Affiliation(s)
- Kaushik Chowdhury
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Shih-Lei Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Rubén Marín-Juez
- Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montreal, QC, Canada.
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
10
|
Grunewald M, Kumar S, Sharife H, Volinsky E, Gileles-Hillel A, Licht T, Permyakova A, Hinden L, Azar S, Friedmann Y, Kupetz P, Tzuberi R, Anisimov A, Alitalo K, Horwitz M, Leebhoff S, Khoma OZ, Hlushchuk R, Djonov V, Abramovitch R, Tam J, Keshet E. Counteracting age-related VEGF signaling insufficiency promotes healthy aging and extends life span. Science 2021; 373:373/6554/eabc8479. [PMID: 34326210 DOI: 10.1126/science.abc8479] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 01/19/2021] [Accepted: 06/06/2021] [Indexed: 12/16/2022]
Abstract
Aging is an established risk factor for vascular diseases, but vascular aging itself may contribute to the progressive deterioration of organ function. Here, we show in aged mice that vascular endothelial growth factor (VEGF) signaling insufficiency, which is caused by increased production of decoy receptors, may drive physiological aging across multiple organ systems. Increasing VEGF signaling prevented age-associated capillary loss, improved organ perfusion and function, and extended life span. Healthier aging was evidenced by favorable metabolism and body composition and amelioration of aging-associated pathologies including hepatic steatosis, sarcopenia, osteoporosis, "inflammaging" (age-related multiorgan chronic inflammation), and increased tumor burden. These results indicate that VEGF signaling insufficiency affects organ aging in mice and suggest that modulating this pathway may result in increased mammalian life span and improved overall health.
Collapse
Affiliation(s)
- M Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - S Kumar
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - H Sharife
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - E Volinsky
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - A Gileles-Hillel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.,Wohl Institute for Translational Medicine and the Goldyne Savad Institute for Gene Therapy, Hadassah Hospital, Jerusalem, Israel.,Pediatric Pulmonology and Sleep Unit, Department of Pediatrics, Hadassah Medical Center, Jerusalem, Israel
| | - T Licht
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - A Permyakova
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - L Hinden
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - S Azar
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Y Friedmann
- Bio-Imaging Unit, The Alexander Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | - P Kupetz
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - R Tzuberi
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - A Anisimov
- Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - K Alitalo
- Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Horwitz
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Leebhoff
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - O Z Khoma
- Topographic and Clinical Anatomy, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - R Hlushchuk
- Topographic and Clinical Anatomy, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - V Djonov
- Topographic and Clinical Anatomy, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - R Abramovitch
- Wohl Institute for Translational Medicine and the Goldyne Savad Institute for Gene Therapy, Hadassah Hospital, Jerusalem, Israel
| | - J Tam
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - E Keshet
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
11
|
Wewers TM, Schulz A, Nolte I, Pavenstädt H, Brand M, Di Marco GS. Circulating Soluble Fms-like Tyrosine Kinase in Renal Diseases Other than Preeclampsia. J Am Soc Nephrol 2021; 32:1853-1863. [PMID: 34155060 PMCID: PMC8455271 DOI: 10.1681/asn.2020111579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Soluble Fms-like tyrosine kinase (sFlt-1/sVEGFR1) is a naturally occurring antagonist of vascular endothelial growth factor (VEGF). Despite being a secreted, soluble protein lacking cytoplasmic and transmembrane domains, sFlt-1 can act locally and be protective against excessive microenvironmental VEGF concentration or exert autocrine functions independently of VEGF. Circulating sFlt-1 may indiscriminately affect endothelial function and the microvasculature of distant target organs. The clinical significance of excess sFlt-1 in kidney disease was first shown in preeclampsia, a major renal complication of pregnancy. However, circulating sFlt-1 levels appear to be increased in various diseases with varying degrees of renal impairment. Relevant clinical associations between circulating sFlt-1 and severe outcomes (e.g., endothelial dysfunction, renal impairment, cardiovascular disease, and all-cause mortality) have been observed in patients with CKD and after kidney transplantation. However, sFlt-1 appears to be protective against renal dysfunction-associated aggravation of atherosclerosis and diabetic nephropathy. Therefore, in this study, we provide an update on sFlt-1 in several kidney diseases other than preeclampsia, discuss clinical findings and experimental studies, and briefly consider its use in clinical practice.
Collapse
Affiliation(s)
- Theresa M. Wewers
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Annika Schulz
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Giovana S. Di Marco
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Correspondence: Giovana S. Di Marco, Albert-Schweitzer-Campus 1, Building A14, 48149 Münster, Germany.
| |
Collapse
|
12
|
Asnani A, Moslehi JJ, Adhikari BB, Baik AH, Beyer AM, de Boer RA, Ghigo A, Grumbach IM, Jain S, Zhu H. Preclinical Models of Cancer Therapy-Associated Cardiovascular Toxicity: A Scientific Statement From the American Heart Association. Circ Res 2021; 129:e21-e34. [PMID: 33934611 DOI: 10.1161/res.0000000000000473] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although cardiovascular toxicity from traditional chemotherapies has been well recognized for decades, the recent explosion of effective novel targeted cancer therapies with cardiovascular sequelae has driven the emergence of cardio-oncology as a new clinical and research field. Cardiovascular toxicity associated with cancer therapy can manifest as a broad range of potentially life-threatening complications, including heart failure, arrhythmia, myocarditis, and vascular events. Beyond toxicology, the intersection of cancer and heart disease has blossomed to include discovery of genetic and environmental risk factors that predispose to both. There is a pressing need to understand the underlying molecular mechanisms of cardiovascular toxicity to improve outcomes in patients with cancer. Preclinical cardiovascular models, ranging from cellular assays to large animals, serve as the foundation for mechanistic studies, with the ultimate goal of identifying biologically sound biomarkers and cardioprotective therapies that allow the optimal use of cancer treatments while minimizing toxicities. Given that novel cancer therapies target specific pathways integral to normal cardiovascular homeostasis, a better mechanistic understanding of toxicity may provide insights into fundamental pathways that lead to cardiovascular disease when dysregulated. The goal of this scientific statement is to summarize the strengths and weaknesses of preclinical models of cancer therapy-associated cardiovascular toxicity, to highlight overlapping mechanisms driving cancer and cardiovascular disease, and to discuss opportunities to leverage cardio-oncology models to address important mechanistic questions relevant to all patients with cardiovascular disease, including those with and without cancer.
Collapse
|
13
|
Dobbin SJ, Petrie MC, Myles RC, Touyz RM, Lang NN. Cardiotoxic effects of angiogenesis inhibitors. Clin Sci (Lond) 2021; 135:71-100. [PMID: 33404052 PMCID: PMC7812690 DOI: 10.1042/cs20200305] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
The development of new therapies for cancer has led to dramatic improvements in survivorship. Angiogenesis inhibitors represent one such advancement, revolutionising treatment for a wide range of malignancies. However, these drugs are associated with cardiovascular toxicities which can impact optimal cancer treatment in the short-term and may lead to increased morbidity and mortality in the longer term. Vascular endothelial growth factor inhibitors (VEGFIs) are associated with hypertension, left ventricular systolic dysfunction (LVSD) and heart failure as well as arterial and venous thromboembolism, QTc interval prolongation and arrhythmia. The mechanisms behind the development of VEGFI-associated LVSD and heart failure likely involve the combination of a number of myocardial insults. These include direct myocardial effects, as well as secondary toxicity via coronary or peripheral vascular damage. Cardiac toxicity may result from the 'on-target' effects of VEGF inhibition or 'off-target' effects resulting from inhibition of other tyrosine kinases. Similar mechanisms may be involved in the development of VEGFI-associated right ventricular (RV) dysfunction. Some VEGFIs can be associated with QTc interval prolongation and an increased risk of ventricular and atrial arrhythmia. Further pre-clinical and clinical studies and trials are needed to better understand the impact of VEGFI on the cardiovascular system. Once mechanisms are elucidated, therapies can be investigated in clinical trials and surveillance strategies for identifying VEGFI-associated cardiovascular complications can be developed.
Collapse
Affiliation(s)
- Stephen J.H. Dobbin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Mark C. Petrie
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rachel C. Myles
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rhian M. Touyz
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Ninian N. Lang
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| |
Collapse
|
14
|
Kunimasa K, Oka T, Hara S, Yamada N, Oizumi S, Miyashita Y, Kamada R, Funamoto T, Kawachi H, Kawamura T, Inoue T, Kuhara H, Tamiya M, Nishino K, Matsunaga T, Imamura F, Fujita M, Kumagai T. Osimertinib is associated with reversible and dose-independent cancer therapy-related cardiac dysfunction. Lung Cancer 2020; 153:186-192. [PMID: 33277055 DOI: 10.1016/j.lungcan.2020.10.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/01/2022]
Abstract
INTRODUCTION The use of osimertinib is associated with the risk of cancer therapy-related cardiac dysfunction (CTRCD) for EGFR-mutated non-small cell lung cancer (NSCLC) patients. In this study, we aimed to clarify the clinical features of patients with CTRCD associated with osimertinib. METHODS A total of 183 cases of advanced EGFR-mutated NSCLC who received osimertinib monotherapy from January 2014 to December 2019 were evaluated. Longitudinal changes in LVEF were evaluated in 58 patients by serial echocardiography before and after osimertinib administration. RESULTS Of 58 patients, 16 patients (8.7%) had decreased LVEF of 10 units or more and 8 patients (4.4%) met the CTRCD criteria. Overall, LVEF significantly decreased after osimertinib treatment from a mean value of 69% (range, 52-82%) at baseline to 66% (26-75%) (p < 0.001). During osimertinib treatment, LVEF remained low but did not decline any further. Discontinuation, dose reduction, or switching to another EGFR tyrosine kinase inhibitors resulted in recovery in 6 out of 8 CTRCD patients. Multivariate analysis showed that history of heart disease was a significant predictor of CTRCD (ORR, 4.97; 95% confidence interval [CI], 1.26-19.6; P = 0.022). CONCLUSIONS Osimertinib was associated with the risk of CTRCD, which is dose-independent and reversible with drug withdrawal.
Collapse
Affiliation(s)
- Kei Kunimasa
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan.
| | - Toru Oka
- Department of Onco-Cardiology, Osaka International Cancer Institute, Osaka, Japan
| | - Satoshi Hara
- Department of Respiratory Medicine, Itami City Hospital, Itami, Japan
| | - Noriyuki Yamada
- Department of Respiratory Medicine, National Hospital Organization Hokkaido Cancer Center, Sapporo, Hokkaido, Japan
| | - Satoshi Oizumi
- Department of Respiratory Medicine, National Hospital Organization Hokkaido Cancer Center, Sapporo, Hokkaido, Japan
| | - Yoshihiro Miyashita
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, Yamanashi, Japan
| | - Risa Kamada
- Department of Onco-Cardiology, Osaka International Cancer Institute, Osaka, Japan
| | - Tomoya Funamoto
- Department of Respiratory Medicine, Itami City Hospital, Itami, Japan
| | - Hayato Kawachi
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Takahisa Kawamura
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Takako Inoue
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hanako Kuhara
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Motohiro Tamiya
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazumi Nishino
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Takashi Matsunaga
- Department of Medical Informatics, Osaka International Cancer Institute, Osaka, Japan
| | - Fumio Imamura
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Masashi Fujita
- Department of Onco-Cardiology, Osaka International Cancer Institute, Osaka, Japan
| | - Toru Kumagai
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
15
|
Campia U, Moslehi JJ, Amiri-Kordestani L, Barac A, Beckman JA, Chism DD, Cohen P, Groarke JD, Herrmann J, Reilly CM, Weintraub NL. Cardio-Oncology: Vascular and Metabolic Perspectives: A Scientific Statement From the American Heart Association. Circulation 2019; 139:e579-e602. [PMID: 30786722 DOI: 10.1161/cir.0000000000000641] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardio-oncology has organically developed as a new discipline within cardiovascular medicine as a result of the cardiac and vascular adverse sequelae of the major advances in cancer treatment. Patients with cancer and cancer survivors are at increased risk of vascular disease for a number of reasons. First, many new cancer therapies, including several targeted therapies, are associated with vascular and metabolic complications. Second, cancer itself serves as a risk factor for vascular disease, especially by increasing the risk for thromboembolic events. Finally, recent data suggest that common modifiable and genetic risk factors predispose to both malignancies and cardiovascular disease. Vascular complications in patients with cancer represent a new challenge for the clinician and a new frontier for research and investigation. Indeed, vascular sequelae of novel targeted therapies may provide insights into vascular signaling in humans. Clinically, emerging challenges are best addressed by a multidisciplinary approach in which cardiovascular medicine specialists and vascular biologists work closely with oncologists in the care of patients with cancer and cancer survivors. This novel approach realizes the goal of providing superior care through the creation of cardio-oncology consultative services and the training of a new generation of cardiovascular specialists with a broad understanding of cancer treatments.
Collapse
|
16
|
Gollmann-Tepeköylü C, Lobenwein D, Theurl M, Primessnig U, Lener D, Kirchmair E, Mathes W, Graber M, Pölzl L, An A, Koziel K, Pechriggl E, Voelkl J, Paulus P, Schaden W, Grimm M, Kirchmair R, Holfeld J. Shock Wave Therapy Improves Cardiac Function in a Model of Chronic Ischemic Heart Failure: Evidence for a Mechanism Involving VEGF Signaling and the Extracellular Matrix. J Am Heart Assoc 2019; 7:e010025. [PMID: 30371289 PMCID: PMC6474945 DOI: 10.1161/jaha.118.010025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Mechanical stimulation of acute ischemic myocardium by shock wave therapy (SWT) is known to improve cardiac function by induction of angiogenesis. However, SWT in chronic heart failure is poorly understood. We aimed to study whether mechanical stimulation upon SWT improves heart function in chronic ischemic heart failure by induction of angiogenesis and postnatal vasculogenesis and to dissect underlying mechanisms. Methods and Results SWT was applied in a mouse model of chronic myocardial ischemia. To study effects of SWT on postnatal vasculogenesis, wild‐type mice received bone marrow transplantation from green fluorescence protein donor mice. Underlying mechanisms were elucidated in vitro in endothelial cells and murine aortic rings. Echocardiography and pressure/volume measurements revealed improved left ventricular ejection fraction, myocardial contractility, and diastolic function and decreased myocardial fibrosis after treatment. Concomitantly, numbers of capillaries and arterioles were increased. SWT resulted in enhanced expression of the chemoattractant stromal cell–derived factor 1 in ischemic myocardium and serum. Treatment induced recruitment of bone marrow–derived endothelial cells to the site of injury. In vitro, SWT resulted in endothelial cell proliferation, enhanced survival, and capillary sprouting. The effects were vascular endothelial growth factor receptor 2 and heparan sulfate proteoglycan dependent. Conclusions SWT positively affects heart function in chronic ischemic heart failure by induction of angiogenesis and postnatal vasculogenesis. SWT upregulated pivotal angiogenic and vasculogenic factors in the myocardium in vivo and induced proliferative and anti‐apoptotic effects on endothelial cells in vitro. Mechanistically, these effects depend on vascular endothelial growth factor signaling and heparan sulfate proteoglycans. SWT is a promising treatment option for regeneration of ischemic myocardium.
Collapse
Affiliation(s)
| | | | - Markus Theurl
- 3 Internal Medicine III Medical University of Innsbruck Austria
| | - Uwe Primessnig
- 4 Department of Internal Medicine and Cardiology Charité - Universitätsmedizin Berlin Germany
| | - Daniela Lener
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | - Elke Kirchmair
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | | | - Michael Graber
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | - Leo Pölzl
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | - Angela An
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | | | - Elisabeth Pechriggl
- 1 Division of Clinical and Functional Anatomy Department of Anatomy, Histology and Embryology Medical University of Innsbruck Austria
| | - Jakob Voelkl
- 4 Department of Internal Medicine and Cardiology Charité - Universitätsmedizin Berlin Germany
| | - Patrick Paulus
- 5 Department of Anaesthesiology and Operative Intensive Care Medicine Kepler University Hospital Linz Austria
| | - Wolfgang Schaden
- 6 Ludwig Boltzmann Institute for Experimental and Clinical Traumatology AUVA Research Centre Vienna Austria.,7 Austrian Cluster for Tissue Regeneration Vienna Austria
| | - Michael Grimm
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | | | - Johannes Holfeld
- 2 Cardiac Surgery Medical University of Innsbruck Austria.,7 Austrian Cluster for Tissue Regeneration Vienna Austria
| |
Collapse
|
17
|
Liu C, Peng M, Zheng L, Zhao Y, Wang R, Su Q, Chen S, Li Z. Enhanced autophagy alleviates injury during hindlimb ischemia/reperfusion in mice. Exp Ther Med 2019; 18:1669-1676. [PMID: 31410124 PMCID: PMC6676216 DOI: 10.3892/etm.2019.7743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 06/06/2019] [Indexed: 12/25/2022] Open
Abstract
Previous studies examining whether autophagy has a protective or deleterious role during ischemia/reperfusion (I/R) injury have reported a varying role in different organs and remains a matter of debate. The aim of the current study was to explore the role of autophagy in hindlimb I/R injury in a murine model. An increase in apoptosis was observed in vitro, in C2C12 myoblast cells, following hypoxia/reoxygenation (H/R), while downregulation of autophagic flux was induced by chloroquine as compared with the vehicle group under hypoxia and H/R conditions. In vivo, an increase in severe damage of gastrocnemius muscles was observed in the I/R and ischemia groups compared with the control group, was more severe in the I/R group compared with the ischemia group. Electron microscopy revealed an increased number of autophagosomes in the ischemia group, whereas a reduced number was detected in the I/R group. Following administration of rapamycin, the infarct size ratio and cell apoptosis was significantly reduced, while the amount of autophagosomes significantly increased in the ischemic phase. In conclusion, autophagy is upregulated in the ischemia phase and downregulated in the reperfusion phase. Notably, upregulation of autophagy via rapamycin intervention during ischemia alleviated skeletal muscle damage, suggesting a potential protective role during hindlimb I/R injury.
Collapse
Affiliation(s)
- Chenshu Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Meixiu Peng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Liang Zheng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yang Zhao
- Division of Interventional Radiology and Vascular Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Rui Wang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qiao Su
- Animal Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Sifan Chen
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
18
|
Tocchetti CG, Cadeddu C, Di Lisi D, Femminò S, Madonna R, Mele D, Monte I, Novo G, Penna C, Pepe A, Spallarossa P, Varricchi G, Zito C, Pagliaro P, Mercuro G. From Molecular Mechanisms to Clinical Management of Antineoplastic Drug-Induced Cardiovascular Toxicity: A Translational Overview. Antioxid Redox Signal 2019; 30:2110-2153. [PMID: 28398124 PMCID: PMC6529857 DOI: 10.1089/ars.2016.6930] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: Antineoplastic therapies have significantly improved the prognosis of oncology patients. However, these treatments can bring to a higher incidence of side-effects, including the worrying cardiovascular toxicity (CTX). Recent Advances: Substantial evidence indicates multiple mechanisms of CTX, with redox mechanisms playing a key role. Recent data singled out mitochondria as key targets for antineoplastic drug-induced CTX; understanding the underlying mechanisms is, therefore, crucial for effective cardioprotection, without compromising the efficacy of anti-cancer treatments. Critical Issues: CTX can occur within a few days or many years after treatment. Type I CTX is associated with irreversible cardiac cell injury, and it is typically caused by anthracyclines and traditional chemotherapeutics. Type II CTX is generally caused by novel biologics and more targeted drugs, and it is associated with reversible myocardial dysfunction. Therefore, patients undergoing anti-cancer treatments should be closely monitored, and patients at risk of CTX should be identified before beginning treatment to reduce CTX-related morbidity. Future Directions: Genetic profiling of clinical risk factors and an integrated approach using molecular, imaging, and clinical data may allow the recognition of patients who are at a high risk of developing chemotherapy-related CTX, and it may suggest methodologies to limit damage in a wider range of patients. The involvement of redox mechanisms in cancer biology and anticancer treatments is a very active field of research. Further investigations will be necessary to uncover the hallmarks of cancer from a redox perspective and to develop more efficacious antineoplastic therapies that also spare the cardiovascular system.
Collapse
Affiliation(s)
| | - Christian Cadeddu
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Daniela Di Lisi
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Saveria Femminò
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- 5 Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy.,6 Department of Internal Medicine, The Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Donato Mele
- 7 Cardiology Unit, Emergency Department, University Hospital of Ferrara, Ferrara, Italy
| | - Ines Monte
- 8 Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Claudia Penna
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Alessia Pepe
- 9 U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Spallarossa
- 10 Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Gilda Varricchi
- 1 Department of Translational Medical Sciences, Federico II University, Naples, Italy.,11 Center for Basic and Clinical Immunology Research (CISI) - Federico II University, Naples, Italy
| | - Concetta Zito
- 12 Division of Cardiology, Clinical and Experimental Department of Medicine and Pharmacology, Policlinico "G. Martino" University of Messina, Messina, Italy
| | - Pasquale Pagliaro
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giuseppe Mercuro
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
19
|
Gogiraju R, Bochenek ML, Schäfer K. Angiogenic Endothelial Cell Signaling in Cardiac Hypertrophy and Heart Failure. Front Cardiovasc Med 2019; 6:20. [PMID: 30895179 PMCID: PMC6415587 DOI: 10.3389/fcvm.2019.00020] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells are, by number, one of the most abundant cell types in the heart and active players in cardiac physiology and pathology. Coronary angiogenesis plays a vital role in maintaining cardiac vascularization and perfusion during physiological and pathological hypertrophy. On the other hand, a reduction in cardiac capillary density with subsequent tissue hypoxia, cell death and interstitial fibrosis contributes to the development of contractile dysfunction and heart failure, as suggested by clinical as well as experimental evidence. Although the molecular causes underlying the inadequate (with respect to the increased oxygen and energy demands of the hypertrophied cardiomyocyte) cardiac vascularization developing during pathological hypertrophy are incompletely understood. Research efforts over the past years have discovered interesting mediators and potential candidates involved in this process. In this review article, we will focus on the vascular changes occurring during cardiac hypertrophy and the transition toward heart failure both in human disease and preclinical models. We will summarize recent findings in transgenic mice and experimental models of cardiac hypertrophy on factors expressed and released from cardiomyocytes, pericytes and inflammatory cells involved in the paracrine (dys)regulation of cardiac angiogenesis. Moreover, we will discuss major signaling events of critical angiogenic ligands in endothelial cells and their possible disturbance by hypoxia or oxidative stress. In this regard, we will particularly highlight findings on negative regulators of angiogenesis, including protein tyrosine phosphatase-1B and tumor suppressor p53, and how they link signaling involved in cell growth and metabolic control to cardiac angiogenesis. Besides endothelial cell death, phenotypic conversion and acquisition of myofibroblast-like characteristics may also contribute to the development of cardiac fibrosis, the structural correlate of cardiac dysfunction. Factors secreted by (dysfunctional) endothelial cells and their effects on cardiomyocytes including hypertrophy, contractility and fibrosis, close the vicious circle of reciprocal cell-cell interactions within the heart during pathological hypertrophy remodeling.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Magdalena L Bochenek
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Katrin Schäfer
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| |
Collapse
|
20
|
Abassi Z, Rosen S, Lamothe S, Heyman SN. Why Have Detection, Understanding and Management of Kidney Hypoxic Injury Lagged Behind those for the Heart? J Clin Med 2019; 8:E267. [PMID: 30795640 PMCID: PMC6406359 DOI: 10.3390/jcm8020267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/17/2019] [Accepted: 02/19/2019] [Indexed: 12/27/2022] Open
Abstract
The outcome of patients with acute myocardial infarction (AMI) has dramatically improved over recent decades, thanks to early detection and prompt interventions to restore coronary blood flow. In contrast, the prognosis of patients with hypoxic acute kidney injury (AKI) remained unchanged over the years. Delayed diagnosis of AKI is a major reason for this discrepancy, reflecting the lack of symptoms and diagnostic tools indicating at real time altered renal microcirculation, oxygenation, functional derangement and tissue injury. New tools addressing these deficiencies, such as biomarkers of tissue damage are yet far less distinctive than myocardial biomarkers and advanced functional renal imaging technologies are non-available in the clinical practice. Moreover, our understanding of pathogenic mechanisms likely suffers from conceptual errors, generated by the extensive use of the wrong animal model, namely warm ischemia and reperfusion. This model parallels mechanistically type I AMI, which properly represents the rare conditions leading to renal infarcts, whereas common scenarios leading to hypoxic AKI parallel physiologically type II AMI, with tissue hypoxic damage generated by altered oxygen supply/demand equilibrium. Better understanding the pathogenesis of hypoxic AKI and its management requires a more extensive use of models of type II-rather than type I hypoxic AKI.
Collapse
Affiliation(s)
- Zaid Abassi
- Department of Physiology, Ruth & Bruce Rappaport Faculty of Medicine, Technion-IIT, Haifa, 31096, Israel.
- Department of Laboratory Medicine, Rambam Health Care campus, Haifa, 31096, Israel.
| | - Seymour Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| | - Simon Lamothe
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| | - Samuel N Heyman
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus, Jerusalem, 91240, Israel.
| |
Collapse
|
21
|
Redfors B, Chen S, Crowley A, Ben-Yehuda O, Gersh BJ, Lembo NJ, Brown WM, Banning AP, Taggart DP, Serruys PW, Kappetein AP, Sabik JF, Stone GW. B-Type Natriuretic Peptide Assessment in Patients Undergoing Revascularization for Left Main Coronary Artery Disease. Circulation 2018; 138:469-478. [DOI: 10.1161/circulationaha.118.033631] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
Elevated B-type natriuretic peptide (BNP) is reflective of impaired cardiac function and is associated with worse prognosis among patients with coronary artery disease (CAD). We sought to assess the association between baseline BNP, adverse outcomes, and the relative efficacy of percutaneous coronary intervention (PCI) versus coronary artery bypass grafting (CABG) in patients with left main CAD.
Methods:
The EXCEL trial (Evaluation of XIENCE Versus Coronary Artery Bypass Surgery for Effectiveness of Left Main Revascularization) randomized patients with left main CAD and low or intermediate SYNTAX scores (Synergy Between PCI With TAXUS and Cardiac Surgery) to PCI with everolimus-eluting stents versus CABG. The primary end point was the composite of all-cause death, myocardial infarction, or stroke. We used multivariable Cox proportional hazards regression to assess the associations between normal versus elevated BNP (≥100 pg/mL), randomized treatment, and the 3-year risk of adverse events.
Results:
BNP at baseline was elevated in 410 of 1037 (39.5%) patients enrolled in EXCEL. Patients with elevated BNP levels were older and more frequently had additional cardiovascular risk factors and lower left ventricular ejection fraction than those with normal BNP, but had similar SYNTAX scores. Patients with elevated BNP had significantly higher 3-year rates of the primary end point (18.6% versus 11.7%; adjusted hazard ratio [HR], 1.62; 95% confidence interval [CI], 1.16–2.28;
P
=0.005) and higher mortality (11.5% versus 3.9%; adjusted HR, 2.49; 95% CI, 1.48–4.19;
P
=0.0006), both from cardiovascular and noncardiovascular causes. In contrast, there were no significant differences in the risks of myocardial infarction, stroke, ischemia-driven revascularization, stent thrombosis, graft occlusion, or major bleeding. A significant interaction (
P
interaction
=0.03) was present between elevated versus normal BNP and treatment with PCI versus CABG for the adjusted risk of the primary composite end point at 3 years among patients with elevated BNP (adjusted HR for PCI versus CABG, 1.54; 95% CI, 0.96–2.47) versus normal BNP (adjusted HR, 0.74; 95% CI, 0.46–1.20). This interaction was stronger when log(BNP) was modeled as a continuous variable (
P
interaction
=0.002).
Conclusions:
In the EXCEL trial, elevated baseline BNP levels in patients with left main CAD undergoing revascularization were independently associated with long-term mortality but not nonfatal adverse ischemic or bleeding events. The relative long-term outcomes after PCI versus CABG for revascularization of left main CAD may be conditioned by the baseline BNP level.
Clinical Trial Registration:
URL:
https://www.clinicaltrials.gov
. Unique identifier: NCT01205776.
Collapse
Affiliation(s)
- Björn Redfors
- Clinical Trials Center, Cardiovascular Research Foundation, New York, NY (B.R., S.C., A.C., O.B.-Y., G.W.S.)
| | - Shmuel Chen
- Clinical Trials Center, Cardiovascular Research Foundation, New York, NY (B.R., S.C., A.C., O.B.-Y., G.W.S.)
| | - Aaron Crowley
- Clinical Trials Center, Cardiovascular Research Foundation, New York, NY (B.R., S.C., A.C., O.B.-Y., G.W.S.)
| | - Ori Ben-Yehuda
- Clinical Trials Center, Cardiovascular Research Foundation, New York, NY (B.R., S.C., A.C., O.B.-Y., G.W.S.)
- New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY (O.B.-Y., N.J.L., G.W.S)
| | - Bernard J. Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Rochester, MN (B.J.G.)
| | - Nicholas J. Lembo
- New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY (O.B.-Y., N.J.L., G.W.S)
| | | | | | - David P. Taggart
- John Radcliffe Hospital, Oxford, United Kingdom (A.P.B., D.P.T.)
| | - Patrick W. Serruys
- Imperial College of Science, Technology and Medicine, London, United Kingdom (P.W.S.)
| | | | - Joseph F. Sabik
- Department of Surgery, University Hospitals Cleveland Medical Center, OH (J.F.S.)
| | - Gregg W. Stone
- Clinical Trials Center, Cardiovascular Research Foundation, New York, NY (B.R., S.C., A.C., O.B.-Y., G.W.S.)
- New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY (O.B.-Y., N.J.L., G.W.S)
| |
Collapse
|
22
|
Broughton KM, Wang BJ, Firouzi F, Khalafalla F, Dimmeler S, Fernandez-Aviles F, Sussman MA. Mechanisms of Cardiac Repair and Regeneration. Circ Res 2018; 122:1151-1163. [PMID: 29650632 PMCID: PMC6191043 DOI: 10.1161/circresaha.117.312586] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular regenerative therapies are pursued on both basic and translational levels. Although efficacy and value of cell therapy for myocardial regeneration can be debated, there is a consensus that profound deficits in mechanistic understanding limit advances, optimization, and implementation. In collaboration with the TACTICS (Transnational Alliance for Regenerative Therapies in Cardiovascular Syndromes), this review overviews several pivotal aspects of biological processes impinging on cardiac maintenance, repair, and regeneration. The goal of summarizing current mechanistic understanding is to prompt innovative directions for fundamental studies delineating cellular reparative and regenerative processes. Empowering myocardial regenerative interventions, whether dependent on endogenous processes or exogenously delivered repair agents, ultimately depends on mastering mechanisms and novel strategies that take advantage of rather than being limited by inherent myocardial biology.
Collapse
Affiliation(s)
- Kathleen M Broughton
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Bingyan J Wang
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Fareheh Firouzi
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Farid Khalafalla
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Stefanie Dimmeler
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Francisco Fernandez-Aviles
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Mark A Sussman
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.).
| |
Collapse
|
23
|
Zelt JGE, Liu PP, Erthal F, deKemp RA, Wells G, O'Meara E, Garrard L, Beanlands RSB, Mielniczuk LM. N-Terminal Pro B-Type Natriuretic Peptide and High-Sensitivity Cardiac Troponin T Levels Are Related to the Extent of Hibernating Myocardium in Patients With Ischemic Heart Failure. Can J Cardiol 2017; 33:1478-1488. [PMID: 28966019 DOI: 10.1016/j.cjca.2017.06.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/07/2017] [Accepted: 06/20/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Increased N-terminal pro b-type natriuretic peptide (NT-proBNP) and high-sensitivity cardiac troponin T (hs-cTnT) can identify patients with heart failure (HF) who are at increased risk of cardiac events. The relationship of these biomarkers to the extent of hibernating myocardium and scar has not been previously characterized in patients with ischemic left ventricular dysfunction and HF. METHODS Patients with ischemic HF meeting recruitment criteria and undergoing perfusion and fluorodeoxyglucose-positron emission tomography to define myocardial hibernation and scar were included in the study. A total of 39 patients (mean age 67 ± 8 years) with New York Heart Association class II-IV HF and ischemic cardiomyopathy (ejection fraction [EF], 27.9% ± 8.5%) were enrolled in the study. RESULTS Serum NT-proBNP and hs-cTnT levels were elevated in patients with ≥ 10% hibernating myocardium compared with those with < 10% (NT-pro-BNP, 7419.10 ± 7169.5 pg/mL vs 2894.6 ± 2967.4 pg/mL; hs-cTnT, 789.3 ± 1835.3 pg/mL vs 44.8 ± 78.9 pg/mL; P < 0.05). The overall receiver operating characteristic under the curve value for NT-proBNP and hs-cTnT to predict hibernating myocardium was 0.76 and 0.78, respectively (P < 0.05). The NT-proBNP (P = 0.02) and hs-cTnT (P < 0.0001) levels also correlated with hibernation, particularly in patients with ≥ 10% scar, independent of EF, age, and estimated glomerular filtration rate. No differences were noted in biomarker levels for patients with vs those without ≥ 10% scar. CONCLUSIONS NT-proBNP and hs-cTnT levels are elevated in patients with ischemic HF hibernation and are correlated with the degree of hibernation but not with the presence or extent of scar. Taken together, these data support the novel concept that NT-proBNP and hs-cTnT release in patients with ischemic HF reflects the presence and extent of hibernating myocardium.
Collapse
Affiliation(s)
- Jason G E Zelt
- Molecular Function and Imaging Program, The National Cardiac PET Centre, and the Advanced Heart Disease Program, Division of Cardiology, Department of Medicine, and the Cardiac Research Methods Centre, University of Ottawa Heart Institute and University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Peter P Liu
- Molecular Function and Imaging Program, The National Cardiac PET Centre, and the Advanced Heart Disease Program, Division of Cardiology, Department of Medicine, and the Cardiac Research Methods Centre, University of Ottawa Heart Institute and University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Fernanda Erthal
- Molecular Function and Imaging Program, The National Cardiac PET Centre, and the Advanced Heart Disease Program, Division of Cardiology, Department of Medicine, and the Cardiac Research Methods Centre, University of Ottawa Heart Institute and University of Ottawa, Ottawa, Ontario, Canada
| | - Robert A deKemp
- Molecular Function and Imaging Program, The National Cardiac PET Centre, and the Advanced Heart Disease Program, Division of Cardiology, Department of Medicine, and the Cardiac Research Methods Centre, University of Ottawa Heart Institute and University of Ottawa, Ottawa, Ontario, Canada
| | - George Wells
- Molecular Function and Imaging Program, The National Cardiac PET Centre, and the Advanced Heart Disease Program, Division of Cardiology, Department of Medicine, and the Cardiac Research Methods Centre, University of Ottawa Heart Institute and University of Ottawa, Ottawa, Ontario, Canada
| | - Eileen O'Meara
- Division of Cardiology, Montréal Heart Institute, Montréal, Québec, Canada
| | - Linda Garrard
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Rob S B Beanlands
- Molecular Function and Imaging Program, The National Cardiac PET Centre, and the Advanced Heart Disease Program, Division of Cardiology, Department of Medicine, and the Cardiac Research Methods Centre, University of Ottawa Heart Institute and University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Lisa M Mielniczuk
- Molecular Function and Imaging Program, The National Cardiac PET Centre, and the Advanced Heart Disease Program, Division of Cardiology, Department of Medicine, and the Cardiac Research Methods Centre, University of Ottawa Heart Institute and University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
24
|
Staels W, Heremans Y, Leuckx G, Van Gassen N, Salinno C, De Groef S, Cools M, Keshet E, Dor Y, Heimberg H, De Leu N. Conditional islet hypovascularisation does not preclude beta cell expansion during pregnancy in mice. Diabetologia 2017; 60:1051-1056. [PMID: 28299380 DOI: 10.1007/s00125-017-4243-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/23/2017] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Endothelial-endocrine cell interactions and vascular endothelial growth factor (VEGF)-A signalling are deemed essential for maternal islet vascularisation, glucose control and beta cell expansion during mouse pregnancy. The aim of this study was to assess whether pregnancy-associated beta cell expansion was affected under conditions of islet hypovascularisation. METHODS Soluble fms-like tyrosine kinase 1 (sFLT1), a VEGF-A decoy receptor, was conditionally overexpressed in maternal mouse beta cells from 1.5 to 14.5 days post coitum. Islet vascularisation, glycaemic control, beta cell proliferation, individual beta cell size and total beta cell volume were assessed in both pregnant mice and non-pregnant littermates. RESULTS Conditional overexpression of sFLT1 in beta cells resulted in islet hypovascularisation and glucose intolerance in both pregnant and non-pregnant mice. In contrast to non-pregnant littermates, glucose intolerance in pregnant mice was transient. sFLT1 overexpression did not affect pregnancy-associated changes in beta cell proliferation, individual beta cell size or total beta cell volume. CONCLUSIONS/INTERPRETATION Reduced intra-islet VEGF-A signalling results in maternal islet hypovascularisation and impaired glycaemic control but does not preclude beta cell expansion during mouse pregnancy.
Collapse
Affiliation(s)
- Willem Staels
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Department of Paediatrics, Division of Paediatric Endocrinology, Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Yves Heremans
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Gunter Leuckx
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Naomi Van Gassen
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Ciro Salinno
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Sofie De Groef
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Martine Cools
- Department of Paediatrics, Division of Paediatric Endocrinology, Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Eli Keshet
- Department of Developmental Biology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Nico De Leu
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium.
- Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium.
| |
Collapse
|
25
|
Chan MC, Ziegler O, Liu L, Rowe GC, Das S, Otterbein LE, Arany Z. Heme oxygenase and carbon monoxide protect from muscle dystrophy. Skelet Muscle 2016; 6:41. [PMID: 27906108 PMCID: PMC5126804 DOI: 10.1186/s13395-016-0114-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/18/2016] [Indexed: 02/07/2023] Open
Abstract
Background Duchenne muscle dystrophy (DMD) is one of the most common lethal genetic diseases of children worldwide and is 100% fatal. Steroids, the only therapy currently available, are marred by poor efficacy and a high side-effect profile. New therapeutic approaches are urgently needed. Methods Here, we leverage PGC-1α, a powerful transcriptional coactivator known to protect against dystrophy in the mdx murine model of DMD, to search for novel mechanisms of protection against dystrophy. Results We identify heme oxygenase-1 (HO-1) as a potential novel target for the treatment of DMD. Expression of HO-1 is blunted in the muscles from the mdx murine model of DMD, and further reduction of HO-1 by genetic haploinsufficiency worsens muscle damage in mdx mice. Conversely, induction of HO-1 pharmacologically protects against muscle damage. Mechanistically, HO-1 degrades heme into biliverdin, releasing in the process ferrous iron and carbon monoxide (CO). We show that exposure to a safe low dose of CO protects against muscle damage in mdx mice, as does pharmacological treatment with CO-releasing molecules. Conclusions These data identify HO-1 and CO as novel therapeutic agents for the treatment of DMD. Safety profiles and clinical testing of inhaled CO already exist, underscoring the translational potential of these observations.
Collapse
Affiliation(s)
- Mun Chun Chan
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Current address: Cardiovascular Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Olivia Ziegler
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Current address: Cardiovascular Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Laura Liu
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Current address: Cardiovascular Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Glenn C Rowe
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Present Address: Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Saumya Das
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Current address: Cardiovascular Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zoltan Arany
- Cardiovascular Institute and Institute Diabetes Obesity and Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, 11th floor, 3400 Civic Blvd, Philadelphia, 19104, PA, USA.
| |
Collapse
|
26
|
Karathanasis N, Tsamardinos I, Lagani V. omicsNPC: Applying the Non-Parametric Combination Methodology to the Integrative Analysis of Heterogeneous Omics Data. PLoS One 2016; 11:e0165545. [PMID: 27812137 PMCID: PMC5094732 DOI: 10.1371/journal.pone.0165545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 10/13/2016] [Indexed: 12/17/2022] Open
Abstract
Background The advance of omics technologies has made possible to measure several data modalities on a system of interest. In this work, we illustrate how the Non-Parametric Combination methodology, namely NPC, can be used for simultaneously assessing the association of different molecular quantities with an outcome of interest. We argue that NPC methods have several potential applications in integrating heterogeneous omics technologies, as for example identifying genes whose methylation and transcriptional levels are jointly deregulated, or finding proteins whose abundance shows the same trends of the expression of their encoding genes. Results We implemented the NPC methodology within “omicsNPC”, an R function specifically tailored for the characteristics of omics data. We compare omicsNPC against a range of alternative methods on simulated as well as on real data. Comparisons on simulated data point out that omicsNPC produces unbiased / calibrated p-values and performs equally or significantly better than the other methods included in the study; furthermore, the analysis of real data show that omicsNPC (a) exhibits higher statistical power than other methods, (b) it is easily applicable in a number of different scenarios, and (c) its results have improved biological interpretability. Conclusions The omicsNPC function competitively behaves in all comparisons conducted in this study. Taking into account that the method (i) requires minimal assumptions, (ii) it can be used on different studies designs and (iii) it captures the dependences among heterogeneous data modalities, omicsNPC provides a flexible and statistically powerful solution for the integrative analysis of different omics data.
Collapse
Affiliation(s)
- Nestoras Karathanasis
- Institute of Computer Science, Foundation for Research and Technology—Hellas, Heraklion, Greece
| | | | - Vincenzo Lagani
- Department of Computer Science, University of Crete, Heraklion, Greece
- * E-mail:
| |
Collapse
|
27
|
Fast revascularization of the injured area is essential to support zebrafish heart regeneration. Proc Natl Acad Sci U S A 2016; 113:11237-11242. [PMID: 27647901 DOI: 10.1073/pnas.1605431113] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Zebrafish have a remarkable capacity to regenerate their heart. Efficient replenishment of lost tissues requires the activation of different cell types including the epicardium and endocardium. A complex set of processes is subsequently needed to support cardiomyocyte repopulation. Previous studies have identified important determinants of heart regeneration; however, to date, how revascularization of the damaged area happens remains unknown. Here, we show that angiogenic sprouting into the injured area starts as early as 15 h after injury. To analyze the role of vegfaa in heart regeneration, we used vegfaa mutants rescued to adulthood by vegfaa mRNA injections at the one-cell stage. Surprisingly, vegfaa mutants develop coronaries and revascularize after injury. As a possible explanation for these observations, we find that vegfaa mutant hearts up-regulate the expression of potentially compensating genes. Therefore, to overcome the lack of a revascularization phenotype in vegfaa mutants, we generated fish expressing inducible dominant negative Vegfaa. These fish displayed minimal revascularization of the damaged area. In the absence of fast angiogenic revascularization, cardiomyocyte proliferation did not occur, and the heart failed to regenerate, retaining a fibrotic scar. Hence, our data show that a fast endothelial invasion allows efficient revascularization of the injured area, which is necessary to support replenishment of new tissue and achieve efficient heart regeneration. These findings revisit the model where neovascularization is considered to happen concomitant with the formation of new muscle. Our work also paves the way for future studies designed to understand the molecular mechanisms that regulate fast revascularization.
Collapse
|
28
|
Chino H, Amano Y, Yamauchi Y, Matsuda J, Takeda N, Tanaka G, Takai D, Nagase T. Cardiogenic syncope possibly related to bevacizumab-containing combination chemotherapy for advanced non-small cell lung cancer. J Thorac Dis 2016; 8:2646-2650. [PMID: 27747019 DOI: 10.21037/jtd.2016.08.96] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We report the case of a 55-year-old man with stage IV lung adenocarcinoma who received carboplatin-paclitaxel-bevacizumab chemotherapy as second-line therapy. After four cycles of chemotherapy, he experienced syncope with a decrease in blood pressure. Electrocardiography (ECG) revealed atrial fibrillation. Cardiac ultrasonography showed a markedly reduced ejection fraction (45%), with moderate decrease in comparison to that before chemotherapy (66%). Bisoprolol fumarate was initiated, and the conversion to sinus rhythm was detected by ECG 4 days after the syncope. At that time, no improvement in the ejection fraction was detected. Bevacizumab-associated cardiotoxicity was suspected, and bevacizumab maintenance therapy was discontinued, although the chemotherapy achieved a stable disease status based on the Response Evaluation Criteria in Solid Tumors. Two months after bevacizumab cessation, the ejection fraction improved to pretreatment level (62%). To the best of our knowledge, this is the first report on cardiogenic syncope due to left ventricular dysfunction that is most consistent with bevacizumab-associated cardiotoxicity in non-small cell lung cancer (NSCLC). Our results indicate that bevacizumab could lead to cardiotoxicity in patients with NSCLC and suggest the importance of the follow-up cardiac ultrasonography.
Collapse
Affiliation(s)
- Haruka Chino
- Department of Respiratory Medicine, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yosuke Amano
- Department of Respiratory Medicine, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yasuhiro Yamauchi
- Department of Respiratory Medicine, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Jun Matsuda
- Department of Cardiovascular Medicine, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Goh Tanaka
- Department of Respiratory Medicine, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Daiya Takai
- Department of Clinical Laboratory, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, the University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
29
|
Herrmann J, Yang EH, Iliescu CA, Cilingiroglu M, Charitakis K, Hakeem A, Toutouzas K, Leesar MA, Grines CL, Marmagkiolis K. Vascular Toxicities of Cancer Therapies: The Old and the New--An Evolving Avenue. Circulation 2016; 133:1272-89. [PMID: 27022039 DOI: 10.1161/circulationaha.115.018347] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Since the late 1990s, there has been a steady decline in cancer-related mortality, in part related to the introduction of so-called targeted therapies. Intended to interfere with a specific molecular pathway, these therapies have, paradoxically, led to a number of effects off their intended cancer tissue or molecular targets. The latest examples are tyrosine kinase inhibitors targeting the Philadelphia Chromosome mutation product, which have been associated with progressive atherosclerosis and acute vascular events. In addition, agents designed to interfere with the vascular growth factor signaling pathway have vascular side effects ranging from hypertension to arterial events and cardiomyocyte toxicity. Interestingly, the risk of cardiotoxicity with drugs such as trastuzumab is predicted by preexisting cardiovascular risk factors and disease, posing the question of a vascular component to the pathophysiology. The effect on the coronary circulation has been the leading explanation for the cardiotoxicity of 5-fluorouracil and may be the underlying the mechanism of presentation of apical ballooning syndrome with various chemotherapeutic agents. Classical chemotherapeutic agents such as cisplatin, often used in combination with bleomycin and vinca alkaloids, can lead to vascular events including acute coronary thrombosis and may be associated with an increased long-term cardiovascular risk. This review is intended to provide an update on the evolving spectrum of vascular toxicities with cancer therapeutics, particularly as they pertain to clinical practice, and to the conceptualization of cardiovascular diseases, as well. Vascular toxicity with cancer therapy: the old and the new, an evolving avenue.
Collapse
Affiliation(s)
- Joerg Herrmann
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.).
| | - Eric H Yang
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Cezar A Iliescu
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Mehmet Cilingiroglu
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Konstantinos Charitakis
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Abdul Hakeem
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Konstantinos Toutouzas
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Massoud A Leesar
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Cindy L Grines
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Konstantinos Marmagkiolis
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| |
Collapse
|
30
|
Gogiraju R, Schroeter MR, Bochenek ML, Hubert A, Münzel T, Hasenfuss G, Schäfer K. Endothelial deletion of protein tyrosine phosphatase-1B protects against pressure overload-induced heart failure in mice. Cardiovasc Res 2016; 111:204-16. [PMID: 27207947 DOI: 10.1093/cvr/cvw101] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
AIMS Cardiac angiogenesis is an important determinant of heart failure. We examined the hypothesis that protein tyrosine phosphatase (PTP)-1B, a negative regulator of vascular endothelial growth factor (VEGF) receptor-2 activation, is causally involved in the cardiac microvasculature rarefaction during hypertrophy and that deletion of PTP1B in endothelial cells prevents the development of heart failure. METHODS AND RESULTS Cardiac hypertrophy was induced by transverse aortic constriction (TAC) in mice with endothelial-specific deletion of PTP1B (End.PTP1B-KO) and controls (End.PTP1B-WT). Survival up to 20 weeks after TAC was significantly improved in mice lacking endothelial PTP1B. Serial echocardiography revealed a better systolic pump function, less pronounced cardiac hypertrophy, and left ventricular dilation compared with End.PTP1B-WT controls. Histologically, banded hearts from End.PTP1B-KO mice exhibited increased numbers of PCNA-positive, proliferating endothelial cells resulting in preserved cardiac capillary density and improved perfusion as well as reduced hypoxia, apoptotic cell death, and fibrosis. Increased relative VEGFR2 and ERK1/2 phosphorylation and greater eNOS expression were present in the hearts of End.PTP1B-KO mice. The absence of PTP1B in endothelial cells also promoted neovascularization following peripheral ischaemia, and bone marrow transplantation excluded a major contribution of Tie2-positive haematopoietic cells to the improved angiogenesis in End.PTP1B-KO mice. Increased expression of caveolin-1 as well as reduced NADPH oxidase-4 expression, ROS generation and TGFβ signalling were observed and may have mediated the cardioprotective effects of endothelial PTP1B deletion. CONCLUSIONS Endothelial PTP1B deletion improves cardiac VEGF signalling and angiogenesis and protects against chronic afterload-induced heart failure. PTP1B may represent a useful target to preserve cardiac function during hypertrophy.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Marco R Schroeter
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Magdalena L Bochenek
- Center for Cardiology, Department of Cardiology I, University Medical Center Mainz, Mainz, Germany Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Astrid Hubert
- Center for Cardiology, Department of Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Department of Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Katrin Schäfer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany Center for Cardiology, Department of Cardiology I, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
31
|
Mercurio V, Pirozzi F, Lazzarini E, Marone G, Rizzo P, Agnetti G, Tocchetti CG, Ghigo A, Ameri P. Models of Heart Failure Based on the Cardiotoxicity of Anticancer Drugs. J Card Fail 2016; 22:449-58. [PMID: 27103426 DOI: 10.1016/j.cardfail.2016.04.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022]
Abstract
Heart failure (HF) is a complication of oncological treatments that may have dramatic clinical impact. It may acutely worsen a patient's condition or it may present with delayed onset, even years after treatment, when cancer has been cured or is in stable remission. Several studies have addressed the mechanisms of cancer therapy-related HF and some have led to the definition of disease models that hold valid for other and more common types of HF. Here, we review these models of HF based on the cardiotoxicity of antineoplastic drugs and classify them in cardiomyocyte-intrinsic, paracrine, or potentially secondary to effects on cardiac progenitor cells. The first group includes HF resulting from the combination of oxidative stress, mitochondrial dysfunction, and activation of the DNA damage response, which is typically caused by anthracyclines, and HF resulting from deranged myocardial energetics, such as that triggered by anthracyclines and sunitinib. Blockade of the neuregulin-1/ErbB4/ErbB2, vascular endothelial growth factor/vascular endothelial growth factor receptor and platelet-derived growth factor /platelet-derived growth factor receptor pathways by trastuzumab, sorafenib and sunitinib is proposed as paradigm of cancer therapy-related HF associated with alterations of myocardial paracrine pathways. Finally, anthracyclines and trastuzumab are also presented as examples of antitumor agents that induce HF by affecting the cardiac progenitor cell population.
Collapse
Affiliation(s)
- Valentina Mercurio
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Flora Pirozzi
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Edoardo Lazzarini
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Giancarlo Marone
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Giulio Agnetti
- Johns Hopkins University, Cardiology, Baltimore, Maryland; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carlo G Tocchetti
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy.
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| |
Collapse
|
32
|
Bellinger AM, Arteaga CL, Force T, Humphreys BD, Demetri GD, Druker BJ, Moslehi JJ. Cardio-Oncology: How New Targeted Cancer Therapies and Precision Medicine Can Inform Cardiovascular Discovery. Circulation 2016; 132:2248-58. [PMID: 26644247 DOI: 10.1161/circulationaha.115.010484] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardio-oncology (the cardiovascular care of cancer patients) has developed as a new translational and clinical field based on the expanding repertoire of mechanism-based cancer therapies. Although these therapies have changed the natural course of many cancers, several may also lead to cardiovascular complications. Many new anticancer drugs approved over the past decade are "targeted" kinase inhibitors that interfere with intracellular signaling contributing to tumor progression. Unexpected cardiovascular and cardiometabolic effects of patient treatment with these inhibitors have provided unique insights into the role of kinases in human cardiovascular biology. Today, an ever-expanding number of cancer therapies targeting novel kinases and other specific cellular and metabolic pathways are being developed and tested in oncology clinical trials. Some of these drugs may affect the cardiovascular system in detrimental ways and others perhaps in beneficial ways. We propose that the numerous ongoing oncology clinical trials are an opportunity for closer collaboration between cardiologists and oncologists to study the cardiovascular and cardiometabolic changes caused by the modulation of these pathways in patients. In this regard, cardio-oncology represents an opportunity and a novel platform for basic and translational investigation and can serve as a potential avenue for optimization of anticancer therapies and for cardiovascular research and drug discovery.
Collapse
Affiliation(s)
- Andrew M Bellinger
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Carlos L Arteaga
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Thomas Force
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Benjamin D Humphreys
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - George D Demetri
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Brian J Druker
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Javid J Moslehi
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.).
| |
Collapse
|
33
|
Heyman SN, Leibowitz D, Mor-Yosef Levi I, Liberman A, Eisenkraft A, Alcalai R, Khamaisi M, Rosenberger C. Adaptive response to hypoxia and remote ischaemia pre-conditioning: a new hypoxia-inducible factors era in clinical medicine. Acta Physiol (Oxf) 2016; 216:395-406. [PMID: 26449466 DOI: 10.1111/apha.12613] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/01/2015] [Accepted: 10/02/2015] [Indexed: 01/30/2023]
Abstract
Transient ischaemia leads to tolerance to subsequent protracted ischaemia. This 'ischaemia pre-conditioning' results from the induction of numerous protective genes, involved in cell metabolism, proliferation and survival, in antioxidant capacity, angiogenesis, vascular tone and erythropoiesis. Hypoxia-inducible factors (HIF) play a pivotal role in this transcriptional adaptive response. HIF prolyl hydroxylases (PHDs), serving as oxygen sensors, control HIFα degradation. HIF-mediated ischaemic pre-conditioning can be achieved with the administration of PHD inhibitors, with the attenuation of organ injury under various hypoxic and toxic insults. Clinical trials are currently under way, evaluating PHD inhibitors as inducers of erythropoietin. Once their safety is established, their potential use might be further tested in clinical trials in various forms of acute ischaemic and toxic organ damage. Repeated transient limb ischaemia was also found to attenuate ischaemic injury in remote organs. This 'remote ischaemic pre-conditioning' phenomenon (RIP) has been extensively studied recently in small clinical trials, preceding, or in parallel with an abrupt insult, such as myocardial infarction, cardiac surgery or radiocontrast administration. Initial results are promising, suggesting organ protection. Large-scale multi-centre studies are currently under way, evaluating the protective potential of RIP in cardiac surgery, in the management of myocardial infarction and in organ transplantation. The mechanisms of organ protection provided by RIP are poorly understood, but HIF seemingly play a role as well. Thus, Inhibition of HIF degradation with PHD inhibitors, as well as RIP (in part through HIF), might develop into novel clinical interventions in organ protection in the near future.
Collapse
Affiliation(s)
- S. N. Heyman
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - D. Leibowitz
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
- Department of Cardiology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - I. Mor-Yosef Levi
- Department of Nephrology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - A. Liberman
- Department of Neurology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - A. Eisenkraft
- The Research Institute for Military Medicine; The Hebrew University Medical School and the Israeli Defense Force Medical Corps; Jerusalem Israel
| | - R. Alcalai
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
- Department of Cardiology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | | | | |
Collapse
|
34
|
Vascular Endothelial Growth Factor (VEGF) Bioavailability Regulates Angiogenesis and Intestinal Stem and Progenitor Cell Proliferation during Postnatal Small Intestinal Development. PLoS One 2016; 11:e0151396. [PMID: 26978773 PMCID: PMC4792464 DOI: 10.1371/journal.pone.0151396] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/26/2016] [Indexed: 02/06/2023] Open
Abstract
Background Vascular endothelial growth factor (VEGF) is a highly conserved, master regulatory molecule required for endothelial cell proliferation, organization, migration and branching morphogenesis. Podocoryne carnea and drosophila, which lack endothelial cells and a vascular system, express VEGF homologs, indicating potential roles beyond angiogenesis and vasculogenesis. The role of VEGF in the development and homeostasis of the postnatal small intestine is unknown. We hypothesized regulating VEGF bioavailability in the postnatal small intestine would exhibit effects beyond the vasculature and influence epithelial cell stem/progenitor populations. Methods VEGF mutant mice were created that overexpressed VEGF in the brush border of epithelium via the villin promotor following doxycycline treatment. To decrease VEGF bioavailability, sFlt-1 mutant mice were generated that overexpressed the soluble VEGF receptor sFlt-1 upon doxycycline administration in the intestinal epithelium. Mice were analyzed after 21 days of doxycycline administration. Results Increased VEGF expression was confirmed by RT-qPCR and ELISA in the intestine of the VEGF mutants compared to littermates. The VEGF mutant duodenum demonstrated increased angiogenesis and vascular leak as compared to littermate controls. The VEGF mutant duodenum revealed taller villi and increased Ki-67-positive cells in the transit-amplifying zone with reduced Lgr5 expression. The duodenum of sFlt-1 mutants revealed shorter villi and longer crypts with reduced proliferation in the transit-amplifying zone, reduced expression of Dll1, Bmp4 and VE-cadherin, and increased expression of Sox9 and EphB2. Conclusions Manipulating VEGF bioavailability leads to profound effects on not only the intestinal vasculature, but epithelial stem and progenitor cells in the intestinal crypt. Elucidation of the crosstalk between VEGF signaling in the vasculature, mesenchyme and epithelial stem/progenitor cell populations may direct future cell therapies for intestinal dysfunction or disease.
Collapse
|
35
|
Abstract
In patients with pulmonary hypertension (PH), the primary cause of death is right ventricular (RV) failure. Improvement in RV function is therefore one of the most important treatment goals. In order to be able to reverse RV dysfunction and also prevent RV failure, a detailed understanding of the pathobiology of RV failure and the underlying mechanisms concerning the transition from a pressure-overloaded adapted right ventricle to a dilated and failing right ventricle is required. Here, we propose that insufficient RV contractility, myocardial fibrosis, capillary rarefaction, and a disturbed metabolism are important features of a failing right ventricle. Furthermore, an overview is provided about the potential direct RV effects of PH-targeted therapies and the effects of RV-directed medical treatments.
Collapse
Affiliation(s)
- Mariëlle C van de Veerdonk
- Department of Pulmonary Diseases, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Harm J Bogaard
- Department of Pulmonary Diseases, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Norbert F Voelkel
- The Victoria Johnson Pulmonary Research Laboratory, Virginia Commonwealth University, 1220 East Broad Street, Richmond, VA, 23298, USA.
| |
Collapse
|
36
|
|
37
|
Molinaro M, Ameri P, Marone G, Petretta M, Abete P, Di Lisa F, De Placido S, Bonaduce D, Tocchetti CG. Recent Advances on Pathophysiology, Diagnostic and Therapeutic Insights in Cardiac Dysfunction Induced by Antineoplastic Drugs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:138148. [PMID: 26583088 PMCID: PMC4637019 DOI: 10.1155/2015/138148] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/01/2015] [Indexed: 12/28/2022]
Abstract
Along with the improvement of survival after cancer, cardiotoxicity due to antineoplastic treatments has emerged as a clinically relevant problem. Potential cardiovascular toxicities due to anticancer agents include QT prolongation and arrhythmias, myocardial ischemia and infarction, hypertension and/or thromboembolism, left ventricular (LV) dysfunction, and heart failure (HF). The latter is variable in severity, may be reversible or irreversible, and can occur soon after or as a delayed consequence of anticancer treatments. In the last decade recent advances have emerged in clinical and pathophysiological aspects of LV dysfunction induced by the most widely used anticancer drugs. In particular, early, sensitive markers of cardiac dysfunction that can predict this form of cardiomyopathy before ejection fraction (EF) is reduced are becoming increasingly important, along with novel therapeutic and cardioprotective strategies, in the attempt of protecting cardiooncologic patients from the development of congestive heart failure.
Collapse
Affiliation(s)
- Marilisa Molinaro
- Department of Medicine and Health Sciences, University of Molise, 86100 Campobasso, Italy
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Giancarlo Marone
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Mario Petretta
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
- National Researches Council, Neuroscience Institute, University of Padova, 35121 Padova, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Domenico Bonaduce
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| |
Collapse
|
38
|
Xie L, Pi X, Townley-Tilson WHD, Li N, Wehrens XHT, Entman ML, Taffet GE, Mishra A, Peng J, Schisler JC, Meissner G, Patterson C. PHD2/3-dependent hydroxylation tunes cardiac response to β-adrenergic stress via phospholamban. J Clin Invest 2015; 125:2759-71. [PMID: 26075818 DOI: 10.1172/jci80369] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/06/2015] [Indexed: 01/08/2023] Open
Abstract
Ischemic heart disease is the leading cause of heart failure. Both clinical trials and experimental animal studies demonstrate that chronic hypoxia can induce contractile dysfunction even before substantial ventricular damage, implicating a direct role of oxygen in the regulation of cardiac contractile function. Prolyl hydroxylase domain (PHD) proteins are well recognized as oxygen sensors and mediate a wide variety of cellular events by hydroxylating a growing list of protein substrates. Both PHD2 and PHD3 are highly expressed in the heart, yet their functional roles in modulating contractile function remain incompletely understood. Here, we report that combined deletion of Phd2 and Phd3 dramatically decreased expression of phospholamban (PLN), resulted in sustained activation of calcium/calmodulin-activated kinase II (CaMKII), and sensitized mice to chronic β-adrenergic stress-induced myocardial injury. We have provided evidence that thyroid hormone receptor-α (TR-α), a transcriptional regulator of PLN, interacts with PHD2 and PHD3 and is hydroxylated at 2 proline residues. Inhibition of PHDs increased the interaction between TR-α and nuclear receptor corepressor 2 (NCOR2) and suppressed Pln transcription. Together, these observations provide mechanistic insight into how oxygen directly modulates cardiac contractility and suggest that cardiac function could be modulated therapeutically by tuning PHD enzymatic activity.
Collapse
|
39
|
Voelkel NF, Bogaard HJ, Gomez-Arroyo J. The need to recognize the pulmonary circulation and the right ventricle as an integrated functional unit: facts and hypotheses (2013 Grover Conference series). Pulm Circ 2015; 5:81-9. [PMID: 25992273 DOI: 10.1086/679702] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/07/2014] [Indexed: 12/31/2022] Open
Abstract
For many patients with severe pulmonary arterial hypertension, heart failure-and, in particular, right heart failure-is the final chapter of their chronic illness. Targeted therapy for pulmonary hypertension is effective only if the right ventricular ejection fraction is maintained or improved. Because improvement of right heart function and reversal of right heart failure are treatment goals, it is important to investigate the cellular and molecular mechanisms that cause right heart failure. Here, we propose that right ventricular capillary rarefaction is an important hallmark of right heart failure and consider that the "sick lung circulation" and the pressure-overloaded right ventricle constitute a functional unit.
Collapse
Affiliation(s)
- Norbert F Voelkel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Vrije Universiteit (VU) Medical Center, Amsterdam, Netherlands
| | - Jose Gomez-Arroyo
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
40
|
Al-Husseini A, Kraskauskas D, Mezzaroma E, Nordio A, Farkas D, Drake JI, Abbate A, Felty Q, Voelkel NF. Vascular endothelial growth factor receptor 3 signaling contributes to angioobliterative pulmonary hypertension. Pulm Circ 2015; 5:101-16. [PMID: 25992275 DOI: 10.1086/679704] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 10/13/2014] [Indexed: 12/13/2022] Open
Abstract
The mechanisms involved in the development of severe angioobliterative pulmonary arterial hypertension (PAH) are multicellular and complex. Many of the features of human severe PAH, including angioobliteration, lung perivascular inflammation, and right heart failure, are reproduced in the Sugen 5416/chronic hypoxia (SuHx) rat model. Here we address, at first glance, the confusing and paradoxical aspect of the model, namely, that treatment of rats with the antiangiogenic vascular endothelial growth factor (VEGF) receptor 1 and 2 kinase inhibitor, Sugen 5416, when combined with chronic hypoxia, causes angioproliferative pulmonary vascular disease. We postulated that signaling through the unblocked VEGF receptor VEGFR3 (or flt4) could account for some of the pulmonary arteriolar lumen-occluding cell growth. We also considered that Sugen 5416-induced VEGFR1 and VEGFR2 blockade could alter the expression pattern of VEGF isoform proteins. Indeed, in the lungs of SuHx rats we found increased expression of the ligand proteins VEGF-C and VEGF-D as well as enhanced expression of the VEGFR3 protein. In contrast, in the failing right ventricle of SuHx rats there was a profound decrease in the expression of VEGF-B and VEGF-D in addition to the previously described reduction in VEGF-A expression. MAZ51, an inhibitor of VEGFR3 phosphorylation and VEGFR3 signaling, largely prevented the development of angioobliteration in the SuHx model; however, obliterated vessels did not reopen when animals with established PAH were treated with the VEGFR3 inhibitor. Part of the mechanism of vasoobliteration in the SuHx model occurs via VEGFR3. VEGFR1/VEGFR2 inhibition can be initially antiangiogenic by inducing lung vessel endothelial cell apoptosis; however, it can be subsequently angiogenic via VEGF-C and VEGF-D signaling through VEGFR3.
Collapse
Affiliation(s)
- Ayser Al-Husseini
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Donatas Kraskauskas
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eleanora Mezzaroma
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Andrea Nordio
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Daniela Farkas
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jennifer I Drake
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Quentin Felty
- Department of Environmental and Occupational Health, Florida International University, Miami, Florida, USA
| | - Norbert F Voelkel
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
41
|
Abdallah AO, Vallurupalli S, Kunthur A. Pazopanib- and bevacizumab-induced reversible heart failure in a patient with metastatic renal cell carcinoma: A case report. J Oncol Pharm Pract 2015; 22:561-5. [PMID: 25956420 DOI: 10.1177/1078155215585189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Al-Ola Abdallah
- Department of Internal Medicine, Division of Hematology and Oncology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, USA Department of Internal Medicine, Division of Hematology and Oncology, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Srikanth Vallurupalli
- Department of Internal Medicine, Division of Cardiology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, USA Department of Internal Medicine, Division of Cardiology, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Anuradha Kunthur
- Department of Internal Medicine, Division of Hematology and Oncology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, USA Department of Internal Medicine, Division of Hematology and Oncology, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
42
|
Simons M, Alitalo K, Annex BH, Augustin HG, Beam C, Berk BC, Byzova T, Carmeliet P, Chilian W, Cooke JP, Davis GE, Eichmann A, Iruela-Arispe ML, Keshet E, Sinusas AJ, Ruhrberg C, Woo YJ, Dimmeler S. State-of-the-Art Methods for Evaluation of Angiogenesis and Tissue Vascularization: A Scientific Statement From the American Heart Association. Circ Res 2015; 116:e99-132. [PMID: 25931450 DOI: 10.1161/res.0000000000000054] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
43
|
Soluble Flt-1 links microvascular disease with heart failure in CKD. Basic Res Cardiol 2015; 110:30. [PMID: 25893874 DOI: 10.1007/s00395-015-0487-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/29/2015] [Accepted: 04/13/2015] [Indexed: 01/17/2023]
Abstract
Chronic kidney disease (CKD) is associated with an increased risk of heart failure (HF). Elevated plasma concentrations of soluble Flt-1 (sFlt-1) have been linked to cardiovascular disease in CKD patients, but whether sFlt-1 contributes to HF in CKD is still unknown. To provide evidence that concludes a pathophysiological role of sFlt-1 in CKD-associated HF, we measured plasma sFlt-1 concentrations in 586 patients with angiographically documented coronary artery disease and renal function classified according to estimated glomerular filtration rate (eGFR). sFlt-1 concentrations correlated negatively with eGFR and were associated with signs of heart failure, based on New York Heart Association functional class and reduced left ventricular ejection fraction (LVEF), and early mortality. Additionally, rats treated with recombinant sFlt-1 showed a 15 % reduction in LVEF and a 29 % reduction in cardiac output compared with control rats. High sFlt-1 concentrations were associated with a 15 % reduction in heart capillary density (number of vessels/cardiomyocyte) and a 24 % reduction in myocardial blood volume. Electron microscopy and histological analysis revealed mitochondrial damage and interstitial fibrosis in the hearts of sFlt-1-treated, but not control rats. In 5/6-nephrectomised rats, an animal model of CKD, sFlt-1 antagonism with recombinant VEGF121 preserved heart microvasculature and significantly improved heart function. Overall, these findings suggest that a component of cardiovascular risk in CKD patients could be directly attributed to sFlt-1. Assessment of patients with CKD confirmed that sFlt-1 concentrations were inversely correlated with renal function, while studies in rats suggested that sFlt-1 may link microvascular disease with HF in CKD.
Collapse
|
44
|
Xu Q, Jennings NL, Sim K, Chang L, Gao XM, Kiriazis H, Lee YY, Nguyen MN, Woodcock EA, Zhang YY, El-Osta A, Dart AM, Du XJ. Pathological hypertrophy reverses β2-adrenergic receptor-induced angiogenesis in mouse heart. Physiol Rep 2015; 3:3/3/e12340. [PMID: 25780088 PMCID: PMC4393171 DOI: 10.14814/phy2.12340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
β-adrenergic activation and angiogenesis are pivotal for myocardial function but the link between both events remains unclear. The aim of this study was to explore the cardiac angiogenesis profile in a mouse model with cardiomyocyte-restricted overexpression of β2-adrenoceptors (β2-TG), and the effect of cardiac pressure overload. β2-TG mice had heightened cardiac angiogenesis, which was essential for maintenance of the hypercontractile phenotype seen in this model. Relative to controls, cardiomyocytes of β2-TGs showed upregulated expression of vascular endothelial growth factor (VEGF), heightened phosphorylation of cAMP-responsive-element-binding protein (CREB), and increased recruitment of phospho-CREB, CREB-binding protein (CBP), and p300 to the VEGF promoter. However, when hearts were subjected to pressure overload by transverse aortic constriction (TAC), angiogenic signaling in β2-TGs was inhibited within 1 week after TAC. β2-TG hearts, but not controls, exposed to pressure overload for 1–2 weeks showed significant increases from baseline in phosphorylation of Ca2+/calmodulin-dependent kinase II (CaMKIIδ) and protein expression of p53, reduction in CREB phosphorylation, and reduced abundance of phospho-CREB, p300 and CBP recruited to the CREB-responsive element (CRE) site of VEGF promoter. These changes were associated with reduction in both VEGF expression and capillary density. While non-TG mice with TAC developed compensatory hypertrophy, (2-TGs exhibited exaggerated hypertrophic growth at week-1 post-TAC, followed by LV dilatation and reduced fractional shortening measured by serial echocardiography. In conclusion, angiogenesis was enhanced by the cardiomyocyte (2AR/CREB/VEGF signaling pathway. Pressure overload rapidly inhibited this signaling, likely as a consequence of activated CaMKII and p53, leading to impaired angiogenesis and functional decompensation.
Collapse
Affiliation(s)
- Qi Xu
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Nicole L Jennings
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kenneth Sim
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Lisa Chang
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ying Ying Lee
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - My-Nhan Nguyen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - You-Yi Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Assam El-Osta
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Anthony M Dart
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia Alfred Heart Centre, the Alfred Hospital, Melbourne, Victoria, Australia Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Licht T, Dor-Wollman T, Ben-Zvi A, Rothe G, Keshet E. Vessel maturation schedule determines vulnerability to neuronal injuries of prematurity. J Clin Invest 2015; 125:1319-28. [PMID: 25689256 DOI: 10.1172/jci79401] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/02/2015] [Indexed: 01/17/2023] Open
Abstract
Premature birth is a major risk factor for multiple brain pathologies, notably periventricular leukomalacia (PVL), which is distinguished by bilateral necrosis of neural tissue around the ventricles and a sequela of neurological disturbances. The 2 hallmarks of brain pathologies of prematurity are a restricted gestational window of vulnerability and confinement of injury to a specific cerebral region. Here, we examined the proposition that both of these features are determined by the state of blood vessel immaturity. We developed a murine genetic model that allows for inducible and reversible VEGF blockade during brain development. Using this system, we determined that cerebral vessels mature in a centrifugal, wave-like fashion that results in sequential acquisition of a functional blood-brain barrier and exit from a VEGF-dependent phase, with periventricular vessels being the last to mature. This developmental program permitted selective ablation of periventricular vessels via episodic VEGF blockade within a specific, vulnerable gestational window. Enforced collapse of ganglionic eminence vessels and resultant periventricular neural apoptosis resulted in a PVL-like phenotype that recapitulates the primary periventricular lesion, ventricular enlargement, and the secondary cortical deficit in out-migrating GABAergic inhibitory interneurons. These findings provide an animal model that reproduces the temporal and spatial specificities of PVL and indicate that damage to VEGF-dependent, immature periventricular vessels contributes to PVL development.
Collapse
|
46
|
|
47
|
Bayeva M, Sawicki KT, Butler J, Gheorghiade M, Ardehali H. Molecular and cellular basis of viable dysfunctional myocardium. Circ Heart Fail 2014; 7:680-91. [PMID: 25028350 DOI: 10.1161/circheartfailure.113.000912] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Marina Bayeva
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Konrad Teodor Sawicki
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Javed Butler
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Mihai Gheorghiade
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.)
| | - Hossein Ardehali
- From the Division of Cardiology, Department of Medicine, Northwestern University School of Medicine, Chicago, IL (M.B., K.T.S., M.G., H.A.); and Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (J.B.).
| |
Collapse
|
48
|
Recognizing and managing left ventricular dysfunction associated with therapeutic inhibition of the vascular endothelial growth factor signaling pathway. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:335. [PMID: 25099086 DOI: 10.1007/s11936-014-0335-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OPINION STATEMENT Therapeutic inhibition of the vascular endothelial growth factor (VEGF) signaling pathway (VSP) is increasingly employed in the contemporary treatment of many cancer types. VSP inhibitors include the anti-VEGF monoclonal antibody (bevacizumab), soluble VEGF receptors (VEGF Trap), and small molecule tyrosine kinase inhibitors (TKIs) targeting the intracellular kinase domain of VEGF receptors. These agents are associated with cardiovascular toxicities such as hypertension, thrombosis, myocardial ischemia, and left ventricular (LV) dysfunction. Data on VSP inhibitor-associated LV dysfunction are largely limited to retrospective studies. Prospective studies are needed to establish the clinical significance of VSP inhibitor-associated LV dysfunction in the general population. Pre-clinical models of VSP inhibitor-associated LV dysfunction have identified mechanisms of cardiotoxicity and may improve our understanding of the pathophysiology underlying other cardiomyopathies. This review provides an overview of LV dysfunction that can occur in patients treated with VSP inhibitors. Potential strategies for clinical detection and management of this cardiotoxicity are explored, while acknowledging that currently available data specific to VSP-inhibitor LV dysfunction are limited. Avenues for future research are suggested.
Collapse
|
49
|
Smith MA. Lessons learned from adult clinical experience to inform evaluations of VEGF pathway inhibitors in children with cancer. Pediatr Blood Cancer 2014; 61:1497-505. [PMID: 24760743 DOI: 10.1002/pbc.25036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 02/27/2014] [Indexed: 01/07/2023]
Abstract
Agents targeting the vascular endothelial growth factor (VEGF) pathway have been studied in adults with cancer for nearly two decades. It is important to assess the lessons learned from this adult experience and to see how these lessons can help inform pediatric development of agents in this class. The benefit achieved from the use of VEGF pathway targeted agents for adult cancers has primarily been to delay for several months disease progression and less commonly time to death for conditions in which cure is not a reasonable expectation. VEGF pathway targeted agents have shown no efficacy when applied in the adjuvant setting. For adults with advanced cancer, prolongation of survival by 2-3 months is considered an important achievement in some settings. However, the primary goal of pediatric oncology clinical research is to identify treatments that allow children to be cured of their cancer and to grow to adulthood without treatment-induced limitations that lower their quality of survival. An important question for the pediatric oncology research community, pharmaceutical companies, and regulatory agencies to address in planning for future clinical trials is whether existing data support a role for VEGF pathway targeted agents in contributing to a therapeutic pathway to cure for children with cancer.
Collapse
|
50
|
Won YW, Bull DA, Kim SW. Functional polymers of gene delivery for treatment of myocardial infarct. J Control Release 2014; 195:110-9. [PMID: 25076177 DOI: 10.1016/j.jconrel.2014.07.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/18/2014] [Accepted: 07/20/2014] [Indexed: 01/18/2023]
Abstract
Ischemic heart disease is rapidly growing as the common cause of death in the world. It is a disease that occurs as a result of coronary artery stenosis and is caused by the lack of oxygen within cardiac muscles due to an imbalance between oxygen supply and demand. The conventional medical therapy is focused on the use of drug eluting stents, coronary-artery bypass graft surgery and anti-thrombosis. Gene therapy provides great opportunities for treatment of cardiovascular disease. In order for gene therapy to be successful, the development of proper gene delivery systems and hypoxia-regulated gene expression vectors is the most important factors. Several non-viral gene transfer methods have been developed to overcome the safety problems of viral transduction. Some of which include plasmids that regulate gene expression that is controlled by environment specific promoters in the transcriptional or the translational level. This review explores polymeric gene carriers that target the myocardium and hypoxia-inducible vectors, which regulate gene expression in response to hypoxia, and their application in animal myocardial infarction models.
Collapse
Affiliation(s)
- Young-Wook Won
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - David A Bull
- Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|