1
|
DeBerge M, Chaudhary R, Schroth S, Thorp EB. Immunometabolism at the Heart of Cardiovascular Disease. JACC Basic Transl Sci 2023; 8:884-904. [PMID: 37547069 PMCID: PMC10401297 DOI: 10.1016/j.jacbts.2022.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 08/08/2023]
Abstract
Immune cell function among the myocardium, now more than ever, is appreciated to regulate cardiac function and pathophysiology. This is the case for both innate immunity, which includes neutrophils, monocytes, dendritic cells, and macrophages, as well as adaptive immunity, which includes T cells and B cells. This function is fueled by cell-intrinsic shifts in metabolism, such as glycolysis and oxidative phosphorylation, as well as metabolite availability, which originates from the surrounding extracellular milieu and varies during ischemia and metabolic syndrome. Immune cell crosstalk with cardiac parenchymal cells, such as cardiomyocytes and fibroblasts, is also regulated by complex cellular metabolic circuits. Although our understanding of immunometabolism has advanced rapidly over the past decade, in part through valuable insights made in cultured cells, there remains much to learn about contributions of in vivo immunometabolism and directly within the myocardium. Insight into such fundamental cell and molecular mechanisms holds potential to inform interventions that shift the balance of immunometabolism from maladaptive to cardioprotective and potentially even regenerative. Herein, we review our current working understanding of immunometabolism, specifically in the settings of sterile ischemic cardiac injury or cardiometabolic disease, both of which contribute to the onset of heart failure. We also discuss current gaps in knowledge in this context and therapeutic implications.
Collapse
Affiliation(s)
| | | | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
2
|
Shi H, El Kazzi M, Liu Y, Gao A, Schroder AL, Vuong S, Young PA, Rayner BS, Vreden C, King NJC, Witting PK. Multiplex analysis of mass imaging data: Application to the pathology of experimental myocardial infarction. Acta Physiol (Oxf) 2022; 235:e13790. [PMID: 35080155 PMCID: PMC9286669 DOI: 10.1111/apha.13790] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022]
Abstract
Aim Imaging mass cytometry (IMC) affords simultaneous immune‐labelling/imaging of multiple antigens in the same tissue. Methods utilizing multiplex data beyond co‐registration are lacking. This study developed and applied an innovative spatial analysis workflow for multiplex imaging data to IMC data determined from cardiac tissues and revealed the mechanism(s) of neutrophil‐mediated post‐myocardial‐infarction damage. Methods IMC produced multiplex images with various redox/inflammatory markers. The cardiac peri‐infarct zone (PIZ) was determined to be up to 240 µm from the infarct border based on the presence of neutrophils. The tissue region beyond the infarct was defined as the remote area (RA). ImageJ was used to quantify the immunoreactivity. Functional assessments included infarct size, cell necro/apoptosis, total thiol assay and echocardiogram. Results Expression of damage markers decreased in order from the infarct area to PIZ and then RA, reflecting the neutrophil density in the regions. Concentrically spaced “shoreline contour analysis” around the cardiac infarct extending into the PIZ showed that immunoreactivity for damage markers decreased linearly with increasing distance from the infarct, concomitant with a decreasing neutrophil‐myeloperoxidase (MPO) gradient from the infarct to the PIZ. Stratifying by concentric bands around individual MPO+‐signal identified that the immunoreactivity of haem‐oxygenase‐1 (HO‐1) and phosphorylated‐p38 mitogen‐activated protein kinase (pP38) peaked near neutrophils. Furthermore, spatial dependence between neutrophils and markers of cardiac cellular damage was confirmed by nearest‐neighbour distance analysis. Post‐infarction tissue exhibited declined functional parameters that were associated with neutrophil migration from the infarct to PIZ. Conclusion This image‐based quantitative protocol revealed the spatial association and provided potential molecular pathways responsible for neutrophil‐mediated damage post‐infarction.
Collapse
Affiliation(s)
- Han Shi
- Redox Biology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Mary El Kazzi
- Redox Biology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Yuyang Liu
- Redox Biology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Antony Gao
- Redox Biology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Angie L. Schroder
- Redox Biology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Sally Vuong
- The Heart Research Institute Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Pamela A. Young
- Australian Centre for Microscopy & Microanalysis Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Benjamin S. Rayner
- The Heart Research Institute Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Caryn Vreden
- Immunopathology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- Sydney Cytometry Facility and Ramaciotti Facility for Human Systems Biology The University of Sydney Sydney New South Wales Australia
| | - Nicholas J. C. King
- Immunopathology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- Sydney Cytometry Facility and Ramaciotti Facility for Human Systems Biology The University of Sydney Sydney New South Wales Australia
- Marie Bashir Institute for Infectious Disease and Biosecurity The University of Sydney Sydney New South Wales Australia
- The University of Sydney Nano Institute The University of Sydney Sydney New South Wales Australia
| | - Paul K. Witting
- Redox Biology Group Discipline of Pathology Faculty of Medicine and Health Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- The University of Sydney Nano Institute The University of Sydney Sydney New South Wales Australia
| |
Collapse
|
3
|
Li M, Han B, Zhao H, Xu C, Xu D, Sieniawska E, Lin X, Kai G. Biological active ingredients of Astragali Radix and its mechanisms in treating cardiovascular and cerebrovascular diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153918. [PMID: 35104756 DOI: 10.1016/j.phymed.2021.153918] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/12/2021] [Accepted: 12/30/2021] [Indexed: 05/13/2023]
Abstract
BACKGROUND With the rising age of the global population, the incidence rate of cardiovascular and cerebrovascular diseases (CCVDs) is increasing, which causes serious public health burden. The efforts for new therapeutic approaches are still being sought since the treatment effects of existing therapies are not quite satisfactory. Chinese traditional medicine proved to be very efficient in the treatment of CCVDs. Well described and established in Chinese medicine, Astragali Radix, has been commonly administered in the prophylaxis and cure of CCVDs for thousands of years. PURPOSE This review summarized the action mode and mechanisms of Astragali Radix phytochemicals on CCVDs, hoping to provide valuable information for the future application, development and improvement of Astragali Radix as well as CCVDs treatment. METHODS A plenty of literature on biological active ingredients of Astragali Radix used for CCVDs treatment were retrieved from online electronic PubMed and Web of Science databases. RESULTS This review highlighted the effects of five main active components in Astragali Radix including astragaloside Ⅳ, cycloastragenol, astragalus polysaccharide, calycosin-7-O-β-d-glucoside, and calycosin on CCVDs. The mechanisms mainly involved anti-oxidative damage, anti-inflammatory, and antiapoptotic through signaling pathways such as PI3K/Akt, Nrf2/HO-1, and TLR4/NF-κB pathway. In addition, the majority active constituents in AR have no obvious toxic side effects. CONCLUSION The main active components of Astragali Radix, especially AS-IV, have been extensively summarized. It has been proved that Astragali Radix has obvious therapeutic effects on various CCVDs, including myocardial and cerebral ischemia, hypertension, atherosclerosis, cardiac hypertrophy, chronic heart failure. CAG possesses anti-ischemia activity without toxicity, indicating a worthy of further development. However, high-quality clinical and pharmacokinetic studies are required to validate the current studies.
Collapse
Affiliation(s)
- Man Li
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Bing Han
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Huan Zhao
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Chongyi Xu
- Wenling Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Taizhou, Zhejiang, 317500, China
| | - Daokun Xu
- Wenling Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Taizhou, Zhejiang, 317500, China
| | - Elwira Sieniawska
- Department of Natural Products Chemistry, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| | - Xianming Lin
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Guoyin Kai
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, The Third Affiliated Hospital, School of Pharmaceutical Sciences, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
4
|
Jurcau A, Simion A. Neuroinflammation in Cerebral Ischemia and Ischemia/Reperfusion Injuries: From Pathophysiology to Therapeutic Strategies. Int J Mol Sci 2021; 23:14. [PMID: 35008440 PMCID: PMC8744548 DOI: 10.3390/ijms23010014] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Its increasing incidence has led stroke to be the second leading cause of death worldwide. Despite significant advances in recanalization strategies, patients are still at risk for ischemia/reperfusion injuries in this pathophysiology, in which neuroinflammation is significantly involved. Research has shown that in the acute phase, neuroinflammatory cascades lead to apoptosis, disruption of the blood-brain barrier, cerebral edema, and hemorrhagic transformation, while in later stages, these pathways support tissue repair and functional recovery. The present review discusses the various cell types and the mechanisms through which neuroinflammation contributes to parenchymal injury and tissue repair, as well as therapeutic attempts made in vitro, in animal experiments, and in clinical trials which target neuroinflammation, highlighting future therapeutic perspectives.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| | - Aurel Simion
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurorehabilitation Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| |
Collapse
|
5
|
Bryson TD, Harding P. Prostaglandin E2 EP receptors in cardiovascular disease: An update. Biochem Pharmacol 2021; 195:114858. [PMID: 34822808 DOI: 10.1016/j.bcp.2021.114858] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022]
Abstract
This review article provides an update for the role of prostaglandin E2 receptors (EP1, EP2, EP3 and EP4) in cardiovascular disease. Where possible we have reported citations from the last decade although this was not possible for all of the topics covered due to the paucity of publications. The authors have attempted to cover the subjects of ischemia-reperfusion injury, arrhythmias, hypertension, novel protein binding partners of the EP receptors and their pathophysiological significance, and cardiac regeneration. These latter two topics bring studies of the EP receptors into new and exciting areas of research that are just beginning to be explored. Where there is peer-reviewed literature, the authors have placed particular emphasis on clinical studies although these are limited in number.
Collapse
Affiliation(s)
- Timothy D Bryson
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI, United States; Frankel Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pamela Harding
- Hypertension & Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States.
| |
Collapse
|
6
|
May SM, Chiang E, Reyes A, Martir G, Patel A, Karmali S, Patel S, West S, Del Arroyo AG, Gourine AV, Ackland GL. Neuromodulation of innate immunity by remote ischaemic conditioning in humans: Experimental cross-over study. Brain Behav Immun Health 2021; 16:100299. [PMID: 34589791 PMCID: PMC8417773 DOI: 10.1016/j.bbih.2021.100299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 02/04/2023] Open
Abstract
Experimental animal studies on the mechanisms of remote ischaemic conditioning (RIC)-induced cardioprotection against ischaemia/reperfusion injury demonstrate involvement of both neuronal and humoral pathways. Autonomic parasympathetic (vagal) pathways confer organ protection through both direct innervation and/or immunomodulation, but evidence in humans is lacking. During acute inflammation, vagal release of acetylcholine suppresses CD11b expression, a critical β2-integrin regulating neutrophil adhesion to the endothelium and transmigration to sites of injury. Here, we tested the hypothesis that RIC recruits vagal activity in humans and has an anti-inflammatory effect by reducing neutrophil CD11b expression. Participants (age:50 ± 19 years; 53% female) underwent ultrasound-guided injection of local anaesthetic within the brachial plexus before applying 3 × 8 min cycles of brachial artery occlusion using a blood pressure cuff (RICblock). RIC was repeated 6 weeks later without brachial plexus block. Masked analysers quantified vagal activity (heart rate, heart rate variability (HRV)) before, and 10 min after, the last cycle of RIC. RR-interval increased after RIC (reduced heart rate) by 40 ms (95% confidence intervals (95%CI):13–66; n = 17 subjects; P = 0.003). RR-interval did not change after brachial plexus blockade (mean difference: 20 ms (95%CI:-11 to 50); P = 0.19). The high-frequency component of HRV was reduced after RICblock, but remained unchanged after RIC (P < 0.001), indicating that RIC preserved vagal activity. LPS-induced CD16+CD11b+ expression in whole blood (measured by flow cytometry) was reduced by RIC (3615 median fluorescence units (95%CI:475-6754); P = 0.026), compared with 2331 units (95%CI:-3921 to 8582); P = 0.726) after RICblock. These data suggest that in humans RIC recruits vagal cardiac and anti-inflammatory mechanisms via ischaemia/reperfusion-induced activation of sensory nerve fibres that innervate the organ undergoing RIC.
Collapse
Affiliation(s)
- Shaun M May
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Eric Chiang
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Anna Reyes
- University College Hospital NHS Trust, London, UK
| | | | - Amour Patel
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Shamir Karmali
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Sanjiv Patel
- University College Hospital NHS Trust, London, UK
| | - Simeon West
- University College Hospital NHS Trust, London, UK
| | - Ana Gutierrez Del Arroyo
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, UK
| | - Gareth L Ackland
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| |
Collapse
|
7
|
Role of Neutrophils in Cardiac Injury and Repair Following Myocardial Infarction. Cells 2021; 10:cells10071676. [PMID: 34359844 PMCID: PMC8305164 DOI: 10.3390/cells10071676] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/26/2022] Open
Abstract
Neutrophils are first-line responders of the innate immune system. Following myocardial infarction (MI), neutrophils are quickly recruited to the ischemic region, where they initiate the inflammatory response, aiming at cleaning up dead cell debris. However, excessive accumulation and/or delayed removal of neutrophils are deleterious. Neutrophils can promote myocardial injury by releasing reactive oxygen species, granular components, and pro-inflammatory mediators. More recent studies have revealed that neutrophils are able to form extracellular traps (NETs) and produce extracellular vesicles (EVs) to aggravate inflammation and cardiac injury. On the contrary, there is growing evidence showing that neutrophils also exert anti-inflammatory, pro-angiogenic, and pro-reparative effects, thus facilitating inflammation resolution and cardiac repair. In this review, we summarize the current knowledge on neutrophils’ detrimental roles, highlighting the role of recently recognized NETs and EVs, followed by a discussion of their beneficial effects and molecular mechanisms in post-MI cardiac remodeling. In addition, emerging concepts about neutrophil diversity and their modulation of adaptive immunity are discussed.
Collapse
|
8
|
Platelets as drivers of ischemia/reperfusion injury after stroke. Blood Adv 2021; 5:1576-1584. [PMID: 33687431 DOI: 10.1182/bloodadvances.2020002888] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide and, despite reperfusion either via thrombolysis or thrombectomy, stroke patients often suffer from lifelong disabilities. These persistent neurological deficits may be improved by treating the ischemia/reperfusion (I/R) injury that occurs following ischemic stroke. There are currently no approved therapies to treat I/R injury, and thus it is imperative to find new targets to decrease the burden of ischemic stroke and related diseases. Platelets, cell fragments from megakaryocytes, are primarily known for their role in hemostasis. More recently, investigators have studied the nonhemostatic role of platelets in inflammatory pathologies, such as I/R injury after ischemic stroke. In this review, we seek to provide an overview of how I/R can lead to platelet activation and how activated platelets, in turn, can exacerbate I/R injury after stroke. We will also discuss potential mechanisms by which platelets may ameliorate I/R injury.
Collapse
|
9
|
Hally KE, Parker OM, Brunton-O'Sullivan MM, Harding SA, Larsen PD. Linking Neutrophil Extracellular Traps and Platelet Activation: A Composite Biomarker Score for Predicting Outcomes after Acute Myocardial Infarction. Thromb Haemost 2021; 121:1637-1649. [PMID: 33984869 DOI: 10.1055/s-0041-1728763] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Activation of both platelets and neutrophils can contribute to the risk of major adverse cardiovascular events (MACE) following acute myocardial infarction (AMI). Neutrophil extracellular traps (NETs) are an important product of the platelet-neutrophil axis and exaggerate vascular damage in cardiovascular disease. Additionally, activated platelets can drive NETosis and are directly linked to thromboembolic risk. Investigating the combined effect of biomarkers for NETosis and platelet activation represents a novel approach to risk prediction post-AMI. Here, we examined the utility of a composite biomarker score, inclusive of both pathways, for predicting MACE post-AMI. METHODS AND RESULTS In a case-control design, 100 case patients who experienced MACE within 1 year of index admission were matched in a 1:2 ratio with control patients. Serum levels of myeloperoxidase-DNA, neutrophil elastase-DNA, and citrullinated histone H3 were assayed by enzyme-linked immunosorbent assay (ELISA) as markers of NET burden. To measure platelet activation, soluble P-selectin was assayed by ELISA in parallel. Platelet and neutrophil counts were also recorded. Composite biomarker scores, inclusive of biomarkers for NETosis and platelet activation, were assessed using multivariate regression modeling. These composite biomarker scores were independent predictors of 1-year MACE. The strongest association with MACE was observed using a composite of platelet count, soluble P-selectin, and all NET markers (odds ratio: 1.94; 1.16-3.25). CONCLUSION Here, we demonstrate the importance of combining biomarkers of NETosis and platelet activation for risk prediction in patients with AMI. Combining biomarkers from closely linked, but distinct, biological pathways was more effective than utilizing either type of biomarker alone.
Collapse
Affiliation(s)
- Kathryn E Hally
- Department of Surgery and Anaesthesia, The University of Otago, Wellington, New Zealand.,School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand
| | - Olivia M Parker
- Department of Surgery and Anaesthesia, The University of Otago, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand
| | - Morgane M Brunton-O'Sullivan
- Department of Surgery and Anaesthesia, The University of Otago, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand
| | - Scott A Harding
- Wellington Cardiovascular Research Group, Wellington, New Zealand.,Department of Cardiology, Wellington Regional Hospital, Wellington, New Zealand
| | - Peter D Larsen
- Department of Surgery and Anaesthesia, The University of Otago, Wellington, New Zealand.,School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand
| |
Collapse
|
10
|
El-Rashid M, Nguyen-Ngo D, Minhas N, Meijles DN, Li J, Ghimire K, Julovi S, Rogers NM. Repurposing of metformin and colchicine reveals differential modulation of acute and chronic kidney injury. Sci Rep 2020; 10:21968. [PMID: 33319836 PMCID: PMC7738483 DOI: 10.1038/s41598-020-78936-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is a major health problem affecting millions of patients globally. There is no effective treatment for AKI and new therapies are urgently needed. Novel drug development, testing and progression to clinical trials is overwhelmingly expensive. Drug repurposing is a more cost-effective measure. We identified 2 commonly used drugs (colchicine and metformin) that alter inflammatory cell function and signalling pathways characteristic of AKI, and tested them in models of acute and chronic kidney injury to assess therapeutic benefit. We assessed the renoprotective effects of colchicine or metformin in C57BL/6 mice challenged with renal ischemia reperfusion injury (IRI), treated before or after injury. All animals underwent analysis of renal function and biomolecular phenotyping at 24 h, 48 h and 4 weeks after injury. Murine renal tubular epithelial cells were studied in response to in vitro mimics of IRI. Pre-emptive treatment with colchicine or metformin protected against AKI, with lower serum creatinine, improved histological changes and decreased TUNEL staining. Pro-inflammatory cytokine profile and multiple markers of oxidative stress were not substantially different between groups. Metformin augmented expression of multiple autophagic proteins which was reversed by the addition of hydroxychloroquine. Colchicine led to an increase in inflammatory cells within the renal parenchyma. Chronic exposure after acute injury to either therapeutic agent in the context of reduced renal mass did not mitigate the development of fibrosis, with colchicine significantly worsening an ischemic phenotype. These data indicate that colchicine and metformin affect acute and chronic kidney injury differently. This has significant implications for potential drug repurposing, as baseline renal disease must be considered when selecting medication.
Collapse
Affiliation(s)
- Maryam El-Rashid
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Danny Nguyen-Ngo
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Sohel Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia. .,Westmead Clinical Medical School, University of Sydney, Camperdown, NSW, Australia. .,Renal Division, Westmead Hospital, Sydney, NSW, Australia. .,Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
El Kazzi M, Rayner BS, Chami B, Dennis JM, Thomas SR, Witting PK. Neutrophil-Mediated Cardiac Damage After Acute Myocardial Infarction: Significance of Defining a New Target Cell Type for Developing Cardioprotective Drugs. Antioxid Redox Signal 2020; 33:689-712. [PMID: 32517486 PMCID: PMC7475094 DOI: 10.1089/ars.2019.7928] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Significance: Acute myocardial infarction (AMI) is a leading cause of death worldwide. Post-AMI survival rates have increased with the introduction of angioplasty as a primary coronary intervention. However, reperfusion after angioplasty represents a clinical paradox, restoring blood flow to the ischemic myocardium while simultaneously inducing ion and metabolic imbalances that stimulate immune cell recruitment and activation, mitochondrial dysfunction and damaging oxidant production. Recent Advances: Preclinical data indicate that these metabolic imbalances contribute to subsequent heart failure through sustaining local recruitment of inflammatory leukocytes and oxidative stress, cardiomyocyte death, and coronary microvascular disturbances, which enhance adverse cardiac remodeling. Both left ventricular dysfunction and heart failure are strongly linked to inflammation and immune cell recruitment to the damaged myocardium. Critical Issues: Overall, therapeutic anti-inflammatory and antioxidant agents identified in preclinical trials have failed in clinical trials. Future Directions: The versatile neutrophil-derived heme enzyme, myeloperoxidase (MPO), is gaining attention as an important oxidative mediator of reperfusion injury, vascular dysfunction, adverse ventricular remodeling, and atrial fibrillation. Accordingly, there is interest in therapeutically targeting neutrophils and MPO activity in the setting of heart failure. Herein, we discuss the role of post-AMI inflammation linked to myocardial damage and heart failure, describe previous trials targeting inflammation and oxidative stress post-AMI, highlight the potential adverse impact of neutrophil and MPO, and detail therapeutic options available to target MPO clinically in AMI patients.
Collapse
Affiliation(s)
- Mary El Kazzi
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | | | - Belal Chami
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Joanne Marie Dennis
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Shane Ross Thomas
- Department of Pathology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Paul Kenneth Witting
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| |
Collapse
|
12
|
Chen J, Ma Q, King JS, Sun Y, Xu B, Zhang X, Zohrabian S, Guo H, Cai W, Li G, Bruno I, Cooke JP, Wang C, Kontaridis M, Wang DZ, Luo H, Pu WT, Lin Z. aYAP modRNA reduces cardiac inflammation and hypertrophy in a murine ischemia-reperfusion model. Life Sci Alliance 2020; 3:e201900424. [PMID: 31843959 PMCID: PMC6918510 DOI: 10.26508/lsa.201900424] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
Myocardial recovery from ischemia-reperfusion (IR) is shaped by the interaction of many signaling pathways and tissue repair processes, including the innate immune response. We and others previously showed that sustained expression of the transcriptional co-activator yes-associated protein (YAP) improves survival and myocardial outcome after myocardial infarction. Here, we asked whether transient YAP expression would improve myocardial outcome after IR injury. After IR, we transiently activated YAP in the myocardium with modified mRNA encoding a constitutively active form of YAP (aYAP modRNA). Histological studies 2 d after IR showed that aYAP modRNA reduced cardiomyocyte (CM) necrosis and neutrophil infiltration. 4 wk after IR, aYAP modRNA-treated mice had better heart function as well as reduced scar size and hypertrophic remodeling. In cultured neonatal and adult CMs, YAP attenuated H2O2- or LPS-induced CM necrosis. TLR signaling pathway components important for innate immune responses were suppressed by YAP/TEAD1. In summary, our findings demonstrate that aYAP modRNA treatment reduces CM necrosis, cardiac inflammation, and hypertrophic remodeling after IR stress.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/administration & dosage
- Adaptor Proteins, Signal Transducing/genetics
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Cardiomegaly/drug therapy
- Cardiomegaly/etiology
- Cell Survival/drug effects
- Cells, Cultured
- Disease Models, Animal
- Humans
- Injections, Intramuscular
- Mice
- Mice, Inbred C57BL
- Myocardial Reperfusion Injury/complications
- Myocarditis/drug therapy
- Myocarditis/etiology
- Myocardium/immunology
- Myocytes, Cardiac/metabolism
- Neutrophil Infiltration/drug effects
- RNA Editing
- RNA, Messenger/administration & dosage
- RNA, Messenger/genetics
- Transcription Factors/administration & dosage
- Transcription Factors/genetics
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Jinmiao Chen
- Boston Children's Hospital, Boston, MA, USA
- Department of Cardiovascular Surgery and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Ma
- Boston Children's Hospital, Boston, MA, USA
| | | | - Yan Sun
- Masonic Medical Research Institute, Utica, NY, USA
| | - Bing Xu
- Masonic Medical Research Institute, Utica, NY, USA
| | | | | | - Haipeng Guo
- Boston Children's Hospital, Boston, MA, USA
- Department of Critical Care Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China
| | - Wenqing Cai
- Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - Gavin Li
- Boston Children's Hospital, Boston, MA, USA
| | - Ivone Bruno
- Houston Methodist Research Institute, Houston, TX, USA
| | - John P Cooke
- Houston Methodist Research Institute, Houston, TX, USA
| | - Chunsheng Wang
- Department of Cardiovascular Surgery and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | - Hongbo Luo
- Boston Children's Hospital, Boston, MA, USA
| | - William T Pu
- Boston Children's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Zhiqiang Lin
- Boston Children's Hospital, Boston, MA, USA
- Masonic Medical Research Institute, Utica, NY, USA
| |
Collapse
|
13
|
Reimer KA, Tanaka M, Murry CE, Richard VJ, Jennings RB. Evaluation of Free Radical Injury in Myocardium. Toxicol Pathol 2019. [DOI: 10.1177/0192623390004part_105] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Abundant evidence now is available that free radicals are produced in excess when myocardium is reperfused following an episode of ischemia and that free radicals can injure myocytes and endothelial cells. Free radicals may contribute to either reversible or irreversible manifestations of cell injury from ischemia and reperfusion. Several investigators have observed that postischemic contractile dysfunction (myocardial stunning) can be attenuated by a variety of anti-free radical therapies, and there seems to be general agreement that free radical injury contributes to stunning. Whether free radicals are an important cause of lethal myocyte injury (“lethal reperfusion injury”) remains controversial. Using similar interventions and animal models, both positive and negative results have been reported from a growing number of studies done to test the effect of anti-free radical therapies on infarct size. Proposed explanations include differences in: 1) dose of drug and onset or duration of treatment, 2) duration of occlusion or reperfusion, 3) methods of measuring infarct size or area at risk, and 4) failure of some studies to control for baseline variation in the major determinants of infarct size, e.g., collateral blood flow. At present, none of these explanations seems sufficient to resolve the question.
Collapse
Affiliation(s)
- Keith A. Reimer
- Department of Pathology, Duke
University Medical Center, Durham, North Carolina 27710
| | - Masaru Tanaka
- The Third Division of Internal
Medicine, Kyoto University, 54 Kawaracho Shogoin, Sakyoku Kyoto, 606, Japan
| | - Charles E. Murry
- Department of Pathology,
University of Washington School of Medicine, Seattle, Washington 98195
| | - Vincent J. Richard
- Departement de Pharmacologie,
Faculte de Medicine Paris Sud, 63 Rue Gabriel Peri, 94276 Le Kremlin-Bicentre Cedex,
France
| | - Robert B. Jennings
- Department of Pathology, Duke
University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
14
|
Lavine KJ, Pinto AR, Epelman S, Kopecky BJ, Clemente-Casares X, Godwin J, Rosenthal N, Kovacic JC. The Macrophage in Cardiac Homeostasis and Disease: JACC Macrophage in CVD Series (Part 4). J Am Coll Cardiol 2019; 72:2213-2230. [PMID: 30360829 DOI: 10.1016/j.jacc.2018.08.2149] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 12/24/2022]
Abstract
Macrophages are integral components of cardiac tissue and exert profound effects on the healthy and diseased heart. Paradigm shifting studies using advanced molecular techniques have revealed significant complexity within these macrophage populations that reside in the heart. In this final of a 4-part review series covering the macrophage in cardiovascular disease, the authors review the origins, dynamics, cell surface markers, and respective functions of each cardiac macrophage subset identified to date, including in the specific scenarios of myocarditis and after myocardial infarction. Looking ahead, a deeper understanding of the diverse and often dichotomous functions of cardiac macrophages will be essential for the development of targeted therapies to mitigate injury and orchestrate recovery of the diseased heart. Moreover, as macrophages are critical for cardiac healing, they are an emerging focus for therapeutic strategies aimed at minimizing cardiomyocyte death, ameliorating pathological cardiac remodeling, and for treating heart failure and after myocardial infarction.
Collapse
Affiliation(s)
- Kory J Lavine
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri; Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri
| | - Alexander R Pinto
- Baker Heart and Diabetes Research Institute, Melbourne, Australia; Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, Toronto, Ontario, Canada
| | - Benjamin J Kopecky
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xavier Clemente-Casares
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - James Godwin
- The Jackson Laboratory, Bar Harbor, Maine; Mt. Desert Island Biological Laboratory, Bar Harbor, Maine
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, Maine; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
15
|
Sodium Channel Nav1.3 Is Expressed by Polymorphonuclear Neutrophils during Mouse Heart and Kidney Ischemia In Vivo and Regulates Adhesion, Transmigration, and Chemotaxis of Human and Mouse Neutrophils In Vitro. Anesthesiology 2019; 128:1151-1166. [PMID: 29509584 DOI: 10.1097/aln.0000000000002135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Voltage-gated sodium channels generate action potentials in excitable cells, but they have also been attributed noncanonical roles in nonexcitable cells. We hypothesize that voltage-gated sodium channels play a functional role during extravasation of neutrophils. METHODS Expression of voltage-gated sodium channels was analyzed by polymerase chain reaction. Distribution of Nav1.3 was determined by immunofluorescence and flow cytometry in mouse models of ischemic heart and kidney injury. Adhesion, transmigration, and chemotaxis of neutrophils to endothelial cells and collagen were investigated with voltage-gated sodium channel inhibitors and lidocaine in vitro. Sodium currents were examined with a whole cell patch clamp. RESULTS Mouse and human neutrophils express multiple voltage-gated sodium channels. Only Nav1.3 was detected in neutrophils recruited to ischemic mouse heart (25 ± 7%, n = 14) and kidney (19 ± 2%, n = 6) in vivo. Endothelial adhesion of mouse neutrophils was reduced by tetrodotoxin (56 ± 9%, unselective Nav-inhibitor), ICA121431 (53 ± 10%), and Pterinotoxin-2 (55 ± 9%; preferential inhibitors of Nav1.3, n = 10). Tetrodotoxin (56 ± 19%), ICA121431 (62 ± 22%), and Pterinotoxin-2 (59 ± 22%) reduced transmigration of human neutrophils through endothelial cells, and also prevented chemotactic migration (n = 60, 3 × 20 cells). Lidocaine reduced neutrophil adhesion to 60 ± 9% (n = 10) and transmigration to 54 ± 8% (n = 9). The effect of lidocaine was not increased by ICA121431 or Pterinotoxin-2. CONCLUSIONS Nav1.3 is expressed in neutrophils in vivo; regulates attachment, transmigration, and chemotaxis in vitro; and may serve as a relevant target for antiinflammatory effects of lidocaine.
Collapse
|
16
|
Marinković G, Grauen Larsen H, Yndigegn T, Szabo IA, Mares RG, de Camp L, Weiland M, Tomas L, Goncalves I, Nilsson J, Jovinge S, Schiopu A. Inhibition of pro-inflammatory myeloid cell responses by short-term S100A9 blockade improves cardiac function after myocardial infarction. Eur Heart J 2019; 40:2713-2723. [DOI: 10.1093/eurheartj/ehz461] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/20/2019] [Accepted: 07/01/2019] [Indexed: 11/13/2022] Open
Abstract
Abstract
Aims
Neutrophils have both detrimental and beneficial effects in myocardial infarction (MI), but little is known about the underlying pathways. S100A8/A9 is a pro-inflammatory alarmin abundantly expressed in neutrophils that is rapidly released in the myocardium and circulation after myocardial ischaemia. We investigated the role of S100A8/A9 in the innate immune response to MI.
Methods and results
In 524 patients with acute coronary syndrome (ACS), we found that high plasma S100A8/A9 at the time of the acute event was associated with lower left ventricular ejection fraction (EF) at 1-year and increased hospitalization for heart failure (HF) during follow-up. In wild-type C57BL/6 mice with MI induced by permanent coronary artery ligation, treatment with the S100A9 blocker ABR-238901 during the inflammatory phase of the immune response inhibited haematopoietic stem cell proliferation and myeloid cell egression from the bone marrow. The treatment reduced the numbers of neutrophils and monocytes/macrophages in the myocardium, promoted an anti-inflammatory environment, and significantly improved cardiac function compared with MI controls. To mimic the clinical scenario, we further confirmed the effects of the treatment in a mouse model of ischaemia/reperfusion. Compared with untreated mice, 3-day ABR-238901 treatment significantly improved left ventricular EF (48% vs. 35%, P = 0.002) and cardiac output (15.7 vs. 11.1 mL/min, P = 0.002) by Day 21 post-MI.
Conclusion
Short-term S100A9 blockade inhibits inflammation and improves cardiac function in murine models of MI. As an excessive S100A8/A9 release is linked to incident HF, S100A9 blockade might represent a feasible strategy to improve prognosis in ACS patients.
Collapse
Affiliation(s)
- Goran Marinković
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
| | - Helena Grauen Larsen
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
| | - Troels Yndigegn
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
| | - Istvan Adorjan Szabo
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Gheorghe Marinescu str. 38, 540139 Targu-Mures, Romania
| | - Razvan Gheorghita Mares
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Gheorghe Marinescu str. 38, 540139 Targu-Mures, Romania
| | - Lisa de Camp
- DeVos Cardiovascular Research Program, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Matthew Weiland
- DeVos Cardiovascular Research Program, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Lukas Tomas
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart & Vascular Institute, Spectrum Health, 100 Michigan Street NE, Grand Rapids, MI 49503, USA
- Cardiovascular Institute, Stanford Medical School, 265 Campus Drive, Stanford, CA 94305, USA
| | - Alexandru Schiopu
- Department of Clinical Sciences Malmö, Lund University, CRC 91:12, Jan Waldenströms gata 35, SE-214 28, Malmö, Sweden
- Department of Cardiology, Skane University Hospital Malmö, Carl-Bertil Laurells gata 9, SE-214 28 Malmö, Sweden
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu-Mures, Gheorghe Marinescu str. 38, 540139 Targu-Mures, Romania
| |
Collapse
|
17
|
Wang Y, Sano S, Oshima K, Sano M, Watanabe Y, Katanasaka Y, Yura Y, Jung C, Anzai A, Swirski FK, Gokce N, Walsh K. Wnt5a-Mediated Neutrophil Recruitment Has an Obligatory Role in Pressure Overload-Induced Cardiac Dysfunction. Circulation 2019; 140:487-499. [PMID: 31170826 DOI: 10.1161/circulationaha.118.038820] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Although the complex roles of macrophages in myocardial injury are widely appreciated, the function of neutrophils in nonischemic cardiac pathology has received relatively little attention. METHODS To examine the regulation and function of neutrophils in pressure overload-induced cardiac hypertrophy, mice underwent treatment with Ly6G antibody to deplete neutrophils and then were subjected to transverse aortic constriction. RESULTS Neutrophil depletion diminished transverse aortic constriction-induced hypertrophy and inflammation and preserved cardiac function. Myeloid deficiency of Wnt5a, a noncanonical Wnt, suppressed neutrophil infiltration to the hearts of transverse aortic constriction-treated mice and produced a phenotype that was similar to the neutropenic conditions. Conversely, mice overexpressing Wnt5a in myeloid cells displayed greater hypertrophic growth, inflammation, and cardiac dysfunction. Neutrophil depletion reversed the Wnt5a overexpression-induced cardiac pathology and eliminated differences in cardiac parameters between wild-type and myeloid-specific Wnt5a transgenic mice. CONCLUSIONS These findings reveal that Wnt5a-regulated neutrophil infiltration has a critical role in pressure overload-induced heart failure.
Collapse
Affiliation(s)
- Ying Wang
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.).,The First Affiliated Hospital of Chongqing Medical University, People's Republic of China (Y. Wang)
| | - Soichi Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| | - Kosei Oshima
- Molecular Cardiology (K.O., Y.K.), Boston University School of Medicine, MA
| | - Miho Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| | - Yosuke Watanabe
- Whitaker Cardiovascular Institute, and Vascular Biology Section (Y. Watanabe), Boston University School of Medicine, MA
| | | | - Yoshimitsu Yura
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| | - Changhee Jung
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.).,Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea (C.J.)
| | - Atsushi Anzai
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston (A.A., F.K.S.)
| | - Filip K Swirski
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston (A.A., F.K.S.)
| | - Noyan Gokce
- Cardiovascular Medicine (N.G.), Boston University School of Medicine, MA.,Cardiology, Boston Medical Center, MA (N.G.). Dr Watanabe is currently at the Department of Internal Medicine II, University of Yamanashi, Faculty of Medicine, Chuo, Yamanashi, Japan. Dr Katanasaka is currently at the Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Japan. Dr Anzai is currently at the Department of Cardiology, Keio University, School of Medicine, Tokyo, Japan
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| |
Collapse
|
18
|
Kithcart AP, Libby P. Unfriendly Fire From Neutrophils Promiscuously Potentiates Cardiovascular Inflammation. Circ Res 2019; 121:1029-1031. [PMID: 29025754 DOI: 10.1161/circresaha.117.311867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Aaron P Kithcart
- From the Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Peter Libby
- From the Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
19
|
Sinusas AJ. Noninvasive Evaluation of No-Reflow Phenomenon. Circ Cardiovasc Imaging 2018; 11:e008576. [PMID: 30571327 DOI: 10.1161/circimaging.118.008576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational, Research Imaging Center, Yale University School of Medicine, New Haven, CT. Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
20
|
Enzmann G, Kargaran S, Engelhardt B. Ischemia-reperfusion injury in stroke: impact of the brain barriers and brain immune privilege on neutrophil function. Ther Adv Neurol Disord 2018; 11:1756286418794184. [PMID: 30181779 PMCID: PMC6111395 DOI: 10.1177/1756286418794184] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023] Open
Abstract
Reperfusion injury following ischemic stroke is a complex pathophysiological process involving numerous mechanisms ranging from the release of excitatory amino acids and ion disequilibrium to the induction of apoptosis and necrosis, to oxidative stress and inflammation. The migration of neutrophils into the brain parenchyma and release of their abundant proteases are generally considered the main cause of neuronal cell death and acute reperfusion injury following ischemic stroke. Recent findings in experimental and human stroke have challenged this view, as the majority of neutrophils were rather found to accumulate within the neurovascular unit (NVU) and the subarachnoid space (SAS) where they remain separated from the brain parenchyma by the glia limitans. The brain parenchyma is an immune-privileged site that is not readily accessible to immune cells and does not elicit stereotypic adaptive or innate immune responses. Understanding brain immune privilege requires intimate knowledge of its unique anatomy in which the brain barriers, that include the glia limitans, establish compartments that differ remarkably with regard to their accessibility to the immune system. We here propose that the brain immune privilege also extends to an ischemic insult, where the brain parenchyma does not evoke a rapid infiltration of neutrophils as observed in ischemic events in peripheral organs. Rather, neutrophil accumulation in the NVU and SAS could have a potential impact on cerebrospinal fluid (CSF) drainage from the central nervous system (CNS) and thus on edema formation and reperfusion injury after ischemic stroke. Integrating the anatomical and functional implications of the brain immune privilege with the unquestionable role of neutrophils in reperfusion injury is a prerequisite to exploit appropriate strategies for therapeutic interventions aiming to reduce neuronal cell death after ischemic stroke.
Collapse
Affiliation(s)
- Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern 3012, Switzerland
| | - Soghra Kargaran
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern 3012, Switzerland
| |
Collapse
|
21
|
Hally KE, Danielson KM, Larsen PD. Looking to the Future: Spotlight on Emerging Biomarkers for Predicting Cardiovascular Risk. CURR EPIDEMIOL REP 2018. [DOI: 10.1007/s40471-018-0158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
22
|
Chorawala MR, Prakash P, Doddapattar P, Jain M, Dhanesha N, Chauhan AK. Deletion of Extra Domain A of Fibronectin Reduces Acute Myocardial Ischaemia/Reperfusion Injury in Hyperlipidaemic Mice by Limiting Thrombo-Inflammation. Thromb Haemost 2018; 118:1450-1460. [PMID: 29960272 DOI: 10.1055/s-0038-1661353] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Fibronectin splicing variant containing extra domain A (Fn-EDA), which is an endogenous ligand for Toll-like receptor 4 (TLR4), is present in negligible amounts in the plasma of healthy humans, but markedly elevated in patients with co-morbid conditions including diabetes and hyperlipidaemia, which are risk factors for myocardial infarction (MI). Very little is known about the role of Fn-EDA in the pathophysiology of acute MI under these co-morbid conditions. MATERIALS AND METHODS We determined the role of Fn-EDA in myocardial ischaemia/reperfusion (I/R) injury in the hyperlipidaemic apolipoprotein E-deficient (ApoE-/-) mice. Infarct size, plasma cardiac troponin I (cTnI) levels, intravascular thrombosis (CD41-positive), neutrophil infiltration (Ly6 B.2-positive), neutrophil extracellular traps (citrullinated H3-positive) and myocyte apoptosis (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling-positive) were assessed in myocardial I/R injury model (1-hour ischaemia/23 hours of reperfusion). RESULTS Irrespective of gender, Fn-EDA-/-ApoE-/- mice exhibited smaller infarct size and decreased cTnI levels concomitant with reduced post-ischaemic intra-vascular thrombi, neutrophils influx, neutrophil extracellular traps and myocyte apoptosis (p < 0.05 vs. ApoE-/- mice). Genetic deletion of TLR4 attenuated myocardial I/R injury in ApoE-/- mice (p < 0.05 vs. ApoE-/- mice), but did not further reduce in Fn-EDA-/- ApoE-/- mice suggesting that Fn-EDA requires TLR4 to mediate myocardial I/R injury. Bone marrow transplantation experiments revealed that Fn-EDA exacerbates myocardial I/R injury through TLR4 expressed on the haematopoietic cells. Infusion of a specific inhibitor of Fn-EDA, 15 minutes post-reperfusion, into ApoE-/- mice attenuated myocardial I/R injury. CONCLUSION Fn-EDA exacerbates TLR4-dependent myocardial I/R injury by promoting post-ischaemic thrombo-inflammatory response. Targeting Fn-EDA may reduce cardiac damage following coronary artery re-canalization after acute MI.
Collapse
Affiliation(s)
- Mehul R Chorawala
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Prem Prakash
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Prakash Doddapattar
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Manish Jain
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Nirav Dhanesha
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Anil K Chauhan
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
23
|
Inflammation and neutrophil immunosenescence in health and disease: Targeted treatments to improve clinical outcomes in the elderly. Exp Gerontol 2018; 105:70-77. [DOI: 10.1016/j.exger.2017.12.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022]
|
24
|
Zhang X, Zhou C, Zhuang J, Xiao X, Zheng S, Xiong W, Wang W, Wu R. Effects of Leukocyte Depletion on Cardiopulmonary Protection and Inflammation after Valve Surgery. Int J Artif Organs 2018. [DOI: 10.1177/039139881003301106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background To evaluate the effects of leukocyte-depleting filtration on myocardial and pulmonary protection and the inflammatory response in patients undergoing valve surgery. Methods Fifty-two patients who underwent mitral valve or mitral and aortic valve replacement were randomized into two groups with or without a leukocyte-depleting filter during surgery. The filter was used from 10 minutes before the release of the aortic cross-clamp to the end of cardiopulmonary bypass. Results Total leukocyte and neutrophil counts showed a short-term reduction in patients undergoing leukocyte filtration, but there was no significant difference between the two groups during the study. Serum levels of cardiac troponin I were lower than that of the control group (p=0.030). Leukocyte depletion resulted in a significantly higher oxygenation index (p=0.002) and a lower respiratory index (p=0.003) compared with the control group. Serum levels of interleukin-8 were significantly elevated in patients undergoing leukocyte filtration compared with patients without leukocyte filtration (p=0.001). There were no statistically significant differences between the two groups with regards to the concentration of interleukin-6 and TNFα, or the duration of intensive care and hospital stay. Conclusions Leukocyte depletion is associated with improved myocardial and lung protection but does not appear to attenuate the inflammatory response in valve surgery.
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| | - Chengbin Zhou
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| | - Jian Zhuang
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| | - Xuejun Xiao
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| | - Shaoyi Zheng
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| | - Weiping Xiong
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| | - Wenxian Wang
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| | - Ruobin Wu
- Department of Cardiac Surgery and Anesthesiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou - China
| |
Collapse
|
25
|
Hu W, Zhang P, Gu J, Yu Q, Zhang D. NEDD4-1 protects against ischaemia/reperfusion-induced cardiomyocyte apoptosis via the PI3K/Akt pathway. Apoptosis 2018; 22:437-448. [PMID: 27837380 DOI: 10.1007/s10495-016-1326-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Activation of the Akt pathway has been shown to protect the heart from ischaemia/reperfusion (I/R) injury. NEDD4-1 has been shown to positively regulate nuclear trafficking of the activated form of Akt. However, the role of NEDD4-1 in cardiac I/R injury remains to be elucidated. In the present study, Lentiviral vectors were constructed to overexpress or knockdown NEDD4-1 in H9c2 cardiomyocytes subjected to I/R injury or ischemic preconditioning (IPC). The results indicated that NEDD4-1 levels were decreased after I/R and increased after IPC in rat heart tissue and in H9c2 cardiomyocytes. Overexpression of NEDD4-1 activated the Akt pathway and regulated apoptosis-related proteins in H9c2 cardiomyocytes, attenuating SI/R-induced cell apoptosis and caspase 3/7 activities. Furthermore, in vivo overexpression of NEDD4-1 attenuated myocardial apoptosis following myocardial I/R. Our results demonstrated that NEDD4-1 protects the myocardium from I/R induced apoptosis by activating PI3K/Akt signaling.
Collapse
Affiliation(s)
- Wei Hu
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, 201199, China.
| | - Peng Zhang
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, 201199, China
| | - Jun Gu
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, 201199, China
| | - Qiang Yu
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, 201199, China
| | - Dadong Zhang
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, 201199, China
| |
Collapse
|
26
|
Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: a continual challenge. Eur Heart J 2018; 38:774-784. [PMID: 27354052 DOI: 10.1093/eurheartj/ehw224] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022] Open
Abstract
The incidence of ST segment elevation myocardial infarction (STEMI) has decreased over the last two decades in developed countries, but mortality from STEMI despite widespread access to reperfusion therapy is still substantial as is the development of heart failure, particularly among an expanding older population. In developing countries, the incidence of STEMI is increasing and interventional reperfusion is often not available. We here review the pathophysiology of acute myocardial infarction and reperfusion, notably the temporal and spatial evolution of ischaemic and reperfusion injury, the different modes of cell death, and the resulting coronary microvascular dysfunction. We then go on to briefly characterize the cardioprotective phenomena of ischaemic preconditioning, ischaemic postconditioning, and remote ischaemic conditioning and their underlying signal transduction pathways. We discuss in detail the attempts to translate conditioning strategies and drug therapy into the clinical setting. Most attempts have failed so far to reduce infarct size and improve clinical outcomes in STEMI patients, and we discuss potential reasons for such failure. Currently, it appears that remote ischaemic conditioning and a few drugs (atrial natriuretic peptide, exenatide, metoprolol, and esmolol) reduce infarct size, but studies with clinical outcome as primary endpoint are still underway.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, 45122 Essen, Germany
| | - Bernard J Gersh
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
27
|
Dunye Guanxinning Improves Acute Myocardial Ischemia-Reperfusion Injury by Inhibiting Neutrophil Infiltration and Caspase-1 Activity. Mediators Inflamm 2018; 2018:4608017. [PMID: 29674944 PMCID: PMC5838499 DOI: 10.1155/2018/4608017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/16/2017] [Accepted: 12/12/2017] [Indexed: 11/17/2022] Open
Abstract
Acute myocardial infarction is the most serious manifestation of cardiovascular disease, and it is a life-threatening condition. Dunye Guanxinning (DG) is a protective traditional Chinese patent herbal medicine with high clinical efficacy and suitable for the treatment of myocardial infarction. However, the mechanism through which it is beneficial is unclear. In this study, we hypothesized that DG improves acute myocardial ischemia-reperfusion injury by inhibiting neutrophil infiltration and caspase-1 activity. We found that DG administration decreased infarct size and cardiomyocyte apoptosis and improved left ventricular ejection fraction, fractional shortening, end-systolic volume index, end-systolic diameter, and carotid arterial blood flow output in rats. DG administration also improved hemorheological parameters, myocardial damage biomarkers, and oxidative stress indexes. The findings showed that DG administration inhibited neutrophil infiltration and reduced the serum interleukin-1 beta (IL-1β) level and myocardial IL-1β maturation. Moreover, DG administration inhibited caspase-1 activity and activated adenosine monophosphate-activated protein kinase (AMPK) phosphorylation in rat hearts. These results suggested that DG administration inhibits inflammasome activity and IL-1β release through the AMPK pathway. Our findings support the clinical efficacy of DG and partially reveal its mechanism, which is beneficial for understanding the therapeutic effects of this protective traditional Chinese patent drug.
Collapse
|
28
|
Shin EY, Wang L, Zemskova M, Deppen J, Xu K, Strobel F, García AJ, Tirouvanziam R, Levit RD. Adenosine Production by Biomaterial-Supported Mesenchymal Stromal Cells Reduces the Innate Inflammatory Response in Myocardial Ischemia/Reperfusion Injury. J Am Heart Assoc 2018; 7:e006949. [PMID: 29331956 PMCID: PMC5850147 DOI: 10.1161/jaha.117.006949] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/30/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND During myocardial ischemia/reperfusion (MI/R) injury, there is extensive release of immunogenic metabolites that activate cells of the innate immune system. These include ATP and AMP, which upregulate chemotaxis, migration, and effector function of early infiltrating inflammatory cells. These cells subsequently drive further tissue devitalization. Mesenchymal stromal cells (MSCs) are a potential treatment modality for MI/R because of their powerful anti-inflammatory capabilities; however, the manner in which they regulate the acute inflammatory milieu requires further elucidation. CD73, an ecto-5'-nucleotidase, may be critical in regulating inflammation by converting pro-inflammatory AMP to anti-inflammatory adenosine. We hypothesized that MSC-mediated conversion of AMP into adenosine reduces inflammation in early MI/R, favoring a micro-environment that attenuates excessive innate immune cell activation and facilitates earlier cardiac recovery. METHODS AND RESULTS Adult rats were subjected to 30 minutes of MI/R injury. MSCs were encapsulated within a hydrogel vehicle and implanted onto the myocardium. A subset of MSCs were pretreated with the CD73 inhibitor, α,β-methylene adenosine diphosphate, before implantation. Using liquid chromatography/mass spectrometry, we found that MSCs increase myocardial adenosine availability following injury via CD73 activity. MSCs also reduce innate immune cell infiltration as measured by flow cytometry, and hydrogen peroxide formation as measured by Amplex Red assay. These effects were dependent on MSC-mediated CD73 activity. Finally, through echocardiography we found that CD73 activity on MSCs was critical to optimal protection of cardiac function following MI/R injury. CONCLUSIONS MSC-mediated conversion of AMP to adenosine by CD73 exerts a powerful anti-inflammatory effect critical for cardiac recovery following MI/R injury.
Collapse
Affiliation(s)
- Eric Y Shin
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Lanfang Wang
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Marina Zemskova
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
- Department of Otolaryngology, College of Medicine, University of Arizona, Tucson, AZ
| | - Juline Deppen
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Kai Xu
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
- Department of Cardiology, Xiangya Hospital of Central South University, Changsha, China
| | | | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | | | - Rebecca D Levit
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
29
|
Shin B, Cowan DB, Emani SM, Del Nido PJ, McCully JD. Mitochondrial Transplantation in Myocardial Ischemia and Reperfusion Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:595-619. [PMID: 28551809 DOI: 10.1007/978-3-319-55330-6_31] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Ischemic heart disease remains the leading cause of death worldwide. Mitochondria are the power plant of the cardiomyocyte, generating more than 95% of the cardiac ATP. Complex cellular responses to myocardial ischemia converge on mitochondrial malfunction which persists and increases after reperfusion, determining the extent of cellular viability and post-ischemic functional recovery. In a quest to ameliorate various points in pathways from mitochondrial damage to myocardial necrosis, exhaustive pharmacologic and genetic tools have targeted various mediators of ischemia and reperfusion injury and procedural techniques without applicable success. The new concept of replacing damaged mitochondria with healthy mitochondria at the onset of reperfusion by auto-transplantation is emerging not only as potential therapy of myocardial rescue, but as gateway to a deeper understanding of mitochondrial metabolism and function. In this chapter, we explore the mechanisms of mitochondrial dysfunction during ischemia and reperfusion, current developments in the methodology of mitochondrial transplantation, mechanisms of cardioprotection and their clinical implications.
Collapse
Affiliation(s)
- Borami Shin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Douglas B Cowan
- Department of Anesthesiology, Division of Cardiac Anesthesia Research, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Sitaram M Emani
- Division of Cardiovascular Critical Care, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Pedro J Del Nido
- Department of Cardiac Surgery, William E. Ladd Professor of Child Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - James D McCully
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, USA.
| |
Collapse
|
30
|
Dong W, Yang R, Yang J, Yang J, Ding J, Wu H, Zhang J. Resveratrol pretreatment protects rat hearts from ischemia/reperfusion injury partly via a NALP3 inflammasome pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2016; 8:8731-41. [PMID: 26464617 DOI: pmid/26464617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/26/2015] [Indexed: 11/18/2022]
Abstract
Inflammatory responses are key players in myocardial ischemia/reperfusion (I/R) injury. Our previous studies showed that resveratrol alleviated I/R injury in myocardial I/R animal models, but whether the NALP3 inflammasome pathway contributes to the mechanisms remains to be elucidated. In this study, we explored the modulation effect of resveratrol on myocardial I/R-induced inflammatory responses in rats. Myocardial I/R rat animal models were induced by occlusion of the left anterior descending coronary arteries (LADs) for 30 min, followed by 2 h of reperfusion. Resveratrol was administered in different doses (2.5, 5, and 10 mg/kg) at the same time as the onset of reperfusion. The serum concentrations of the trinitrotoluene (TnT) and MB isoenzyme creatine kinase (CK-MB) were detected using an automatic biochemical analyzer. Myocardial ultrastructure and morphology were observed with an electron microscope and a light microscope. Myocardial ischemia and infarct sizes were evaluated using Evans blue and tetrazolium chloride (TTC) staining. The NALP3, Caspase1, interleukin 1β (IL-1β) and interleukin 18 (IL-18) mRNA levels were evaluated using RT-PCR. The NALP3 and Caspase1 protein expression levels were detected by western blotting. The IL-1β and IL-18 content in peripheral blood was measured by enzyme-linked immunosorbent assay (ELISA). The myocardial structure in myocardial ischemia reperfusion injury (MI/RI) rats was extensively damaged. After preconditioning with different concentrations of resveratrol (2.5, 5 and 10 mg/kg), the pathology and morphology were significantly improved in a dose-dependent manner. Our results showed that resveratrol treatment significantly reduced the infarct volume and myocardial fibrosis, resulting in myocardial cells that lined up in a more orderly fashion and dose-dependent decreases in TnT and CK-MB levels in the serum of the I/R rats. Resveratrol also significantly modulated mRNA and protein levels by down-regulating NALP3 and Caspase1 expression and IL-1β and IL-18 activation. These results suggest that the NALP3 inflammasome is activated during the myocardial I/R injury process and that the secretion of the inflammatory cytokines IL-1β and IL-18 mediates the cascade inflammatory response. Resveratrol may play an important role in protecting the myocardium against I/R injury in rats by inhibiting the expression and activation of the NALP3 inflammatory body. Therefore, the attenuation of the inflammatory response may be involved in the cardioprotective mechanisms of resveratrol in response to myocardial I/R injury.
Collapse
Affiliation(s)
- Wusong Dong
- Department of Cardiology, The First College of Clinical Medicine, China Three Gorges University Yichang, Hubei, China
| | - Rui Yang
- Department of Medicine, University of Vermont Burlington, vt, USA ; Tongji Medical College, Huazhong University of Science and Technology Wuhan, Hubei Province, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medicine, China Three Gorges University Yichang, Hubei, China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medicine, China Three Gorges University Yichang, Hubei, China
| | - Jiawang Ding
- Department of Cardiology, The First College of Clinical Medicine, China Three Gorges University Yichang, Hubei, China
| | - Hui Wu
- Department of Cardiology, The First College of Clinical Medicine, China Three Gorges University Yichang, Hubei, China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medicine, China Three Gorges University Yichang, Hubei, China
| |
Collapse
|
31
|
Schloss MJ, Horckmans M, Nitz K, Duchene J, Drechsler M, Bidzhekov K, Scheiermann C, Weber C, Soehnlein O, Steffens S. The time-of-day of myocardial infarction onset affects healing through oscillations in cardiac neutrophil recruitment. EMBO Mol Med 2016; 8:937-48. [PMID: 27226028 PMCID: PMC4967945 DOI: 10.15252/emmm.201506083] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death in Western countries. Epidemiological studies show acute MI to be more prevalent in the morning and to be associated with a poorer outcome in terms of mortality and recovery. The mechanisms behind this association are not fully understood. Here, we report that circadian oscillations of neutrophil recruitment to the heart determine infarct size, healing, and cardiac function after MI. Preferential cardiac neutrophil recruitment during the active phase (Zeitgeber time, ZT13) was paralleled by enhanced myeloid progenitor production, increased circulating numbers of CXCR2hi neutrophils as well as upregulated cardiac adhesion molecule and chemokine expression. MI at ZT13 resulted in significantly higher cardiac neutrophil infiltration compared to ZT5, which was inhibited by CXCR2 antagonism or neutrophil‐specific CXCR2 knockout. Limiting exaggerated neutrophilic inflammation at this time point significantly reduced the infarct size and improved cardiac function.
Collapse
Affiliation(s)
- Maximilian J Schloss
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Michael Horckmans
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Katrin Nitz
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Johan Duchene
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Maik Drechsler
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany Department of Pathology, Amsterdam Medical Center (AMC), Amsterdam, The Netherlands
| | - Kiril Bidzhekov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Christoph Scheiermann
- Walter-Brendel-Center of Experimental Medicine, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany Department of Pathology, Amsterdam Medical Center (AMC), Amsterdam, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
32
|
In Vivo Cardioprotective Effects and Pharmacokinetic Profile of N-Propyl Caffeamide Against Ischemia Reperfusion Injury. Arch Immunol Ther Exp (Warsz) 2016; 65:145-156. [DOI: 10.1007/s00005-016-0413-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/09/2016] [Indexed: 01/01/2023]
|
33
|
Ferrari R, Biscaglia S, Malagù M, Bertini M, Campo G. Can We Improve Myocardial Protection during Ischaemic Injury? Cardiology 2016; 135:14-26. [DOI: 10.1159/000444847] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/19/2016] [Indexed: 11/19/2022]
|
34
|
Jose Corbalan J, Vatner DE, Vatner SF. Myocardial apoptosis in heart disease: does the emperor have clothes? Basic Res Cardiol 2016; 111:31. [PMID: 27043720 DOI: 10.1007/s00395-016-0549-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/24/2016] [Indexed: 01/06/2023]
Abstract
Since the discovery of a novel mechanism of cell death that differs from traditional necrosis, i.e., apoptosis, there have been numerous studies concluding that increased apoptosis augments myocardial infarction and heart failure and that limiting apoptosis protects the heart. Importantly, the vast majority of cells in the heart are non-myocytes with only roughly 30 % myocytes, yet almost the entire field studying apoptosis in the heart has disregarded non-myocyte apoptosis, e.g., only 4.7 % of 423 studies on myocardial apoptosis in the past 3 years quantified non-myocyte apoptosis. Accordingly, we reviewed the history of apoptosis in the heart focusing first on myocyte apoptosis, followed by the history of non-myocyte apoptosis in myocardial infarction and heart failure. Apoptosis of several of the major non-myocyte cell types in the heart (cardiac fibroblasts, endothelial cells, vascular smooth muscle cells, macrophages and leukocytes) may actually be responsible for affecting the severity of myocardial infarction and heart failure. In summary, even though it is now known that the majority of apoptosis in the heart occurs in non-myocytes, very little work has been done to elucidate the mechanisms by which non-myocyte apoptosis might be responsible for the adverse effects of apoptosis in myocardial infarction and heart failure. The goal of this review is to provide an impetus for future work in this field on non-myocyte apoptosis that will be required for a better understanding of the role of apoptosis in the heart.
Collapse
Affiliation(s)
- J Jose Corbalan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
35
|
Modification of Caffeic Acid with Pyrrolidine Enhances Antioxidant Ability by Activating AKT/HO-1 Pathway in Heart. PLoS One 2016; 11:e0148545. [PMID: 26845693 PMCID: PMC4742076 DOI: 10.1371/journal.pone.0148545] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/19/2016] [Indexed: 12/03/2022] Open
Abstract
Overproduction of free radicals during ischemia/reperfusion (I/R) injury leads to an interest in using antioxidant therapy. Activating an endogenous antioxidant signaling pathway is more important due to the fact that the free radical scavenging behavior in vitro does not always correlate with a cytoprotection effect in vivo. Caffeic acid (CA), an antioxidant, is a major phenolic constituent in nature. Pyrrolidinyl caffeamide (PLCA), a derivative of CA, was compared with CA for their antioxidant and cytoprotective effects. Our results indicate that CA and PLCA exert the same ability to scavenge DPPH in vitro. In response to myocardial I/R stress, PLCA was shown to attenuate lipid peroxydation and troponin release more than CA. These responses were accompanied with a prominent elevation in AKT and HO-1 expression and a preservation of mnSOD expression and catalase activity. PLCA also improved cell viability and alleviated the intracellular ROS level more than CA in cardiomyocytes exposed to H2O2. When inhibiting the AKT or HO-1 pathways, PLCA lost its ability to recover mnSOD expression and catalase activity to counteract with oxidative stress, suggesting AKT/HO-1 pathway activation by PLCA plays an important role. In addition, inhibition of AKT signaling further abolished HO-1 activity, while inhibition of HO-1 signaling attenuated AKT expression, indicating cross-talk between the AKT and HO-1 pathways. These protective effects may contribute to the cardiac function improvement by PLCA. These findings provide new insight into therapeutic approaches using a modified natural compound against oxidative stress from myocardial injuries.
Collapse
|
36
|
Zhang M, Pan K, Liu Q, Zhou X, Jiang T, Li Y. Growth differentiation factor 15 may protect the myocardium from no‑reflow by inhibiting the inflammatory‑like response that predominantly involves neutrophil infiltration. Mol Med Rep 2015; 13:623-32. [PMID: 26647773 PMCID: PMC4686086 DOI: 10.3892/mmr.2015.4573] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 08/17/2015] [Indexed: 01/13/2023] Open
Abstract
The aim of the current study was to investigate the time course of the expression of growth differentiation factor‑15 (GDF‑15) in rat ischemic myocardium with increasing durations of reperfusion, and to elucidate its physiopathological role in the no‑reflow phenomenon. Wistar rats were randomly divided into ischemia reperfusion (I/R) and sham groups, and myocardial I/R was established by ligation of the left anterior descending coronary artery for 1 h followed by reperfusion for 2, 4, 6, 12, 24 h and 7 days whilst rats in the sham group were subjected to a sham operation. The expression levels of GDF‑15 and ICAM‑1 were measured, in addition to myeloperoxidase (MPO) activity. The myocardial anatomical no‑reflow and infarction areas were assessed. The area at risk was not significantly different following various periods of reperfusion, while the infarct area and no‑reflow area were significantly greater following 6 h of reperfusion (P<0.05). The mRNA and protein expression levels of GDF‑15 were increased during the onset and development of no‑reflow, and peaked following 24 h of reperfusion. MPO activity was reduced with increasing reperfusion duration, while ICAM‑1 levels were increased. Hematoxylin and eosin staining demonstrated that myocardial neutrophil infiltration was significantly increased by I/R injury, in particular following 2, 4 and 6 h of reperfusion. GDF‑15 expression levels were negatively correlated with MPO activity (r=‑0.55, P<0.001), and the MPO activity was negatively correlated with the area of no‑reflow (r=‑0.46, P<0.01). By contrast, GDF‑15 was significantly positively correlated with ICAM‑1 levels (r=0.52, P<0.01), which additionally were demonstrated to be significantly positively associated with the size of the no‑reflow area (r=0.39, P<0.05). The current study demonstrated the time course effect of reperfusion on the expression of GDF‑15 in the myocardial I/R rat model, with the shorter reperfusion times (6 h) resulting in significant no‑reflow in ischemic myocardium. GDF‑15 may protect the I/R myocardium from no‑reflow by inhibiting the inflammatory‑like response, which involves neutrophil infiltration and transendothelial migration.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Cardiology, The Affiliated Hospital of Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Kunying Pan
- Department of Cardiology, The Affiliated Hospital of Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Qianping Liu
- Department of Cardiology, The Affiliated Hospital of Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Institute of Cardiovascular Disease and Heart Center, Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Tiemin Jiang
- Department of Cardiology, The Affiliated Hospital of Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Yuming Li
- Department of Cardiology, The Affiliated Hospital of Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| |
Collapse
|
37
|
Zhu M, Goetsch SC, Wang Z, Luo R, Hill JA, Schneider J, Morris SM, Liu ZP. FoxO4 promotes early inflammatory response upon myocardial infarction via endothelial Arg1. Circ Res 2015; 117:967-77. [PMID: 26438688 DOI: 10.1161/circresaha.115.306919] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/02/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Inflammation in post-myocardial infarction (MI) is necessary for myocyte repair and wound healing. Unfortunately, it is also a key component of subsequent heart failure pathology. Transcription factor forkhead box O4 (FoxO4) regulates a variety of biological processes, including inflammation. However, its role in MI remains unknown. OBJECTIVE To test the hypothesis that FoxO4 promotes early post-MI inflammation via endothelial arginase 1 (Arg1). METHODS AND RESULTS We induced MI in wild-type and FoxO4(-/-) mice. FoxO4(-/-) mice had a significantly higher post-MI survival, better cardiac function, and reduced infarct size. FoxO4(-/-) hearts had significantly fewer neutrophils, reduced expression of cytokines, and competitive nitric oxide synthase inhibitor Arg1. We generated conditional FoxO4 knockout mice with FoxO4 deleted in cardiac mycoytes or endothelial cells. FoxO4 endothelial cell-specific knockout mice showed significant post-MI improvement of cardiac function and reduction of neutrophil accumulation and cytokine expression, whereas FoxO4 cardiac mycoyte-specific knockout mice had no significant difference in cardiac function and post-MI inflammation from those of control littermates. FoxO4 binds the Foxo-binding site in the Arg1 promoter and activates Arg1 transcription. FoxO4 knockdown in human aortic endothelial cells upregulated nitric oxide on ischemia and suppressed monocyte adhesion that can be reversed by ectopic-expression of Arg1. Furthermore, chemical inhibition of Arg1 in wild-type mice had similar cardioprotection and reduced inflammation after MI as FoxO4 inactivation and administration of nitric oxide synthase inhibitor to FoxO4 KO mice reversed the beneficial effects of FoxO4 deletion on post-MI cardiac function. CONCLUSIONS FoxO4 activates Arg1 transcription in endothelial cells in response to MI, leading to downregulation of nitric oxide and upregulation of neutrophil infiltration to the infarct area.
Collapse
Affiliation(s)
- Min Zhu
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.)
| | - Sean C Goetsch
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.)
| | - Zhaoning Wang
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.)
| | - Robert Luo
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.)
| | - Joseph A Hill
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.)
| | - Jay Schneider
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.)
| | - Sidney M Morris
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.)
| | - Zhi-Ping Liu
- From the Departments of Internal Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX (M.Z., S.C.G., Z.W., R.L., J.A.H., J.S., Z.-P.L.); and Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, PA (S.M.M.).
| |
Collapse
|
38
|
Plant Natural Products Calycosin and Gallic Acid Synergistically Attenuate Neutrophil Infiltration and Subsequent Injury in Isoproterenol-Induced Myocardial Infarction: A Possible Role for Leukotriene B4 12-Hydroxydehydrogenase? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:434052. [PMID: 26265982 PMCID: PMC4523677 DOI: 10.1155/2015/434052] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/16/2015] [Accepted: 06/24/2015] [Indexed: 01/29/2023]
Abstract
Leukotriene B4 12-hydroxydehydrogenase (LTB4DH) catalyzes the oxidation of proinflammatory LTB4 into less bioactive 12-oxo-LTB4. We recently discovered that LTB4DH was induced by two different natural products in combination. We previously isolated gallic acid from Radix Paeoniae through a bioactivity-guided fractionation procedure. The purpose of this study is to test the hypothesis that LTB4DH inducers may suppress neutrophil-mediated inflammation in myocardial infarction. We first isolated the active compound(s) from another plant, Radix Astragali, by the similar strategy. By evaluating LTB4DH induction, we identified calycosin and formononetin from Radix Astragali by HPLC-ESI-MS technique. We confirmed that gallic acid and commercial calycosin or formononetin could synergistically induce LTB4DH expression in HepG2 cells and human neutrophils. Moreover, calycosin and gallic acid attenuated the effects of LTB4 on the survival and chemotaxis of neutrophil cell culture. We further demonstrated that calycosin and gallic acid synergistically suppressed neutrophil infiltration and protected cardiac integrity in the isoproterenol-induced mice model of myocardial infarction. Calycosin and gallic acid dramatically suppressed isoproterenol-induced increase in myeloperoxidase (MPO) activity and malondialdehyde (MDA) level. Collectively, our results suggest that LTB4DH inducers (i.e., calycosin and gallic acid) may be a novel combined therapy for the treatment of neutrophil-mediated myocardial injury.
Collapse
|
39
|
Affiliation(s)
- Ryan A Frieler
- From Department of Molecular and Integrative Physiology (R.A.F., R.M.M.), Department of Internal Medicine, Metabolism, Endocrinology, and Diabetes Division (R.M.M.), and Department of Pharmacology (R.M.M.), University of Michigan Medical School, Ann Arbor
| | - Richard M Mortensen
- From Department of Molecular and Integrative Physiology (R.A.F., R.M.M.), Department of Internal Medicine, Metabolism, Endocrinology, and Diabetes Division (R.M.M.), and Department of Pharmacology (R.M.M.), University of Michigan Medical School, Ann Arbor.
| |
Collapse
|
40
|
Soraya H, Rameshrad M, Mokarizadeh A, Garjani A. Metformin attenuates myocardial remodeling and neutrophil recruitment after myocardial infarction in rat. ACTA ACUST UNITED AC 2015; 5:3-8. [PMID: 25901291 PMCID: PMC4401166 DOI: 10.15171/bi.2015.02] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 09/28/2014] [Accepted: 09/30/2014] [Indexed: 11/18/2022]
Abstract
![]()
Introduction: Acute treatment with metformin has a cardio-protective effects by suppression of inflammatory responses during myocardial infarction (MI) through activation of AMP-activated protein kinase (AMPK). Neutrophils have a pivotal role during MI-induced inflammatory responses. Some anti-inflammatory treatments have decreased cardiac injury and infarct size in MI. Here we evaluated the effects of chronic pre-treatment with metformin on myocardial remodeling and neutrophil recruitment after isoproterenol-induced MI.
Methods: Male wistar rats were randomly assigned into 6 groups (n=6) of untreated control, sham, isoproterenol (Iso), and pre-treated orally with 25, 50, and 100 mg/kg of metformin, twice daily, for 14 days. Isoproterenol was injected subcutaneously (sc) at 13th and 14th days for induction of acute MI. Histopathological examinations were done on the harvested hearts. Number of neutrophils in peripheral blood and their infiltration to myocardium were evaluated by Gimsa staining and myeloperoxidase (MPO) assay, respectively.
Results: Histopathological analysis showed a significant attenuation of isoproterenol-induced cardiomyocyte necrosis and fibrosis by all three doses of metformin. The heart to body weight ratio was also decreased with all doses of metformin. Pre-treatment with metformin in comparison to Iso (MI) group reduced peripheral neutrophils (p<0.05, p<0.01, and p<0.001 at 25, 50, and 100 mg/kg; respectively) as well as MPO activity (p<0.05 and p<0.01 at 50 and 100 mg/ kg, respectively).
Conclusion: Pre-treatment with metformin decreased post-MI myocardial injuries by reducing cardiac remodeling and myocardial neutrophil activity. The results could be explained as a new mechanism for cardio-protective effect of metformin.
Collapse
Affiliation(s)
- Hamid Soraya
- Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Rameshrad
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aram Mokarizadeh
- Cellular and Molecular Research Center, and Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Alireza Garjani
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Ku HC, Lee SY, Chen CH, Wang YH, Lin CT, Lee SS, Li TH, Su MJ. TM-1-1DP exerts protective effect against myocardial ischemia reperfusion injury via AKT-eNOS pathway. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:539-48. [PMID: 25672911 DOI: 10.1007/s00210-015-1098-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/28/2015] [Indexed: 12/28/2022]
Abstract
Coronary heart disease remains a leading cause of death in the world. The demand on targeting therapy to reduce myocardial ischemia/reperfusion (I/R) injury is still urgent. The pathogenesis of I/R-induced myocardial injury is complicated. Reactive oxygen species (ROS) generation and inflammatory response activation participate in the development of I/R injury. Cell death occurs and finally leads to myocardial infarction. A newly phenolic aporphine alkaloid derivative, TM-1-1DP, was synthesized in our team. We aimed to investigate the effect of novel compound on myocardial I/R injury. Rats were subjected to 1-h coronary artery occlusion and followed by 2-h reperfusion. Adult rat cardimoycyte was isolated for the cell study, and H2O2 was added into culture medium to induce ROS stress. As compared to the sham group, TM-1-1DP-treated rats had better cardiac performance in association with less infarct size and cardiac injury markers after myocardial I/R. The protective effect is associated with the inhibition of inflammatory response, cell death-related pathway (caspase-3 and TNF-α), and the activation of AKT-eNOS pathway. The finding was further coincided with the cell study. TM-1-1DP treatment significantly alleviated ROS production and improved cell viability in cardiomyocyte after H2O2 exposure. The action of TM-1-1DP is via a nitric oxide (NO)-dependent manner, since NOS inhibitor, L-NAME, abolished the protective effect. We provide a new insight into this therapeutic potential for phenolic aporphine alkaloid in myocardial I/R.
Collapse
Affiliation(s)
- Hui-Chun Ku
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Qin C, Yang YH, May L, Gao X, Stewart AG, Tu Y, Woodman OL, Ritchie RH. Cardioprotective potential of annexin-A1 mimetics in myocardial infarction. Pharmacol Ther 2014; 148:47-65. [PMID: 25460034 DOI: 10.1016/j.pharmthera.2014.11.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI) and its resultant heart failure remains a major cause of death in the world. The current treatments for patients with MI are revascularization with thrombolytic agents or interventional procedures. These treatments have focused on restoring blood flow to the ischemic tissue to prevent tissue necrosis and preserve organ function. The restoration of blood flow after a period of ischemia, however, may elicit further myocardial damage, called reperfusion injury. Pharmacological interventions, such as antioxidant and Ca(2+) channel blockers, have shown premises in experimental settings; however, clinical studies have shown limited success. Thus, there is a need for the development of novel therapies to treat reperfusion injury. The therapeutic potential of glucocorticoid-regulated anti-inflammatory mediator annexin-A1 (ANX-A1) has recently been recognized in a range of systemic inflammatory disorders. ANX-A1 binds to and activates the family of formyl peptide receptors (G protein-coupled receptor family) to inhibit neutrophil activation, migration and infiltration. Until recently, studies on the cardioprotective actions of ANX-A1 and its peptide mimetics (Ac2-26, CGEN-855A) have largely focused on its anti-inflammatory effects as a mechanism of preserving myocardial viability following I-R injury. Our laboratory provided the first evidence of the direct protective action of ANX-A1 on myocardium, independent of inflammatory cells in vitro. We now review the potential for ANX-A1 based therapeutics to be seen as a "triple shield" therapy against myocardial I-R injury, limiting neutrophil infiltration and preserving both cardiomyocyte viability and contractile function. This novel therapy may thus represent a valuable clinical approach to improve outcome after MI.
Collapse
Affiliation(s)
- Chengxue Qin
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Yuan H Yang
- Centre for Inflammatory Diseases Monash University and Monash Medical Centre, Clayton, Victoria, Australia
| | - Lauren May
- Department of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Xiaoming Gao
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Alastair G Stewart
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Yan Tu
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Owen L Woodman
- School of Medical Sciences, RMIT University, Bundoora 3083, Victoria, Australia
| | - Rebecca H Ritchie
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
43
|
|
44
|
Yan L, Vatner SF, Vatner DE. Disruption of type 5 adenylyl cyclase prevents β-adrenergic receptor cardiomyopathy: a novel approach to β-adrenergic receptor blockade. Am J Physiol Heart Circ Physiol 2014; 307:H1521-8. [PMID: 25193472 DOI: 10.1152/ajpheart.00491.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
β-Adrenergic receptor (β-AR) blockade is widely used to treat heart failure, since the adverse effects of chronic β-AR stimulation are central to the pathogenesis of this disease state. Transgenic (Tg) mice, where β-AR signaling is chronically enhanced by overexpression of cardiac β₂-ARs, is a surrogate for this mechanism, since these mice develop cardiomyopathy as reflected by reduced left ventricular (LV) function, increased fibrosis, apoptosis, and myocyte hypertrophy. We hypothesized that disruption of type 5 adenylyl cyclase (AC5), which is in the β-AR signaling pathway in the heart, but exerts only a minor β-AR blocking effect, could prevent the cardiomyopathy in β₂-AR Tg mice without the negative effects of full β-AR blockade. Accordingly, we mated β₂-AR Tg mice with AC5 knockout (KO) mice. The β₂-AR Tg × AC5 KO bigenic mice prevented the cardiomyopathy as reflected by improved LV ejection fraction, reduced apoptosis, fibrosis, and myocyte size and preserved exercise capacity. The rescue was not simply due to a β-blocking effect of AC5 KO, since neither baseline LV function nor the response to isoproterenol was diminished substantially compared with the negative inotropic effects of β-blockade. However, AC5 disruption in β₂-AR Tg activates the antioxidant, manganese superoxide dismutase, an important mechanism protecting the heart from cardiomyopathy. These results indicate that disruption of AC5 prevents the cardiomyopathy induced by chronically enhanced β-AR signaling in mice with overexpressed β₂-AR, potentially by enhancing resistance to oxidative stress and apoptosis, suggesting a novel, alternative approach to β-AR blockade.
Collapse
Affiliation(s)
- Lin Yan
- Departments of Cell Biology and Molecular Medicine and Medicine and the Cardiovascular Research Institute, Rutgers University-New Jersey Medical School, Newark, New Jersey
| | - Stephen F Vatner
- Departments of Cell Biology and Molecular Medicine and Medicine and the Cardiovascular Research Institute, Rutgers University-New Jersey Medical School, Newark, New Jersey
| | - Dorothy E Vatner
- Departments of Cell Biology and Molecular Medicine and Medicine and the Cardiovascular Research Institute, Rutgers University-New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
45
|
Knorr M, Münzel T, Wenzel P. Interplay of NK cells and monocytes in vascular inflammation and myocardial infarction. Front Physiol 2014; 5:295. [PMID: 25177297 PMCID: PMC4132269 DOI: 10.3389/fphys.2014.00295] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/22/2014] [Indexed: 01/08/2023] Open
Abstract
Inflammatory monocytes and macrophages have been identified as key players in the pathogenesis of atherosclerosis, arterial hypertension, and myocardial infarction (MI). They become powerful mediators of vascular inflammation through their capacity to secrete and induce the production of proinflammatory cytokines, chemokines and adhesion molecules and through the production of reactive oxygen species mainly via their NADPH oxidase. Importantly, a crosstalk exists between NK cells and monocytes that works via a feedforwad amplification loop of T-bet/Interferon-gamma/interleukin-12 signaling, that causes mutual activation of both NK cells and monocytes and that fosters recruitment of inflammatory cells to sites of inflammation. Recently, we have discovered that this crosstalk is crucial for the unrestricted development of angiotensin II (ATII) induced vascular injury in arterial hypertension, the most important risk factor for atherosclerosis and cardiovascular disease worldwide. In this review, we will also discuss possible implications of this interplay between NK cells and monocytes for the pathogenesis of coronary atherosclerosis and myocardial infarction and potential therapeutic options.
Collapse
Affiliation(s)
- Maike Knorr
- Department of Medicine 2 and Center for Thrombosis and Hemostasis University Medical Center Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Medicine 2 and Center for Thrombosis and Hemostasis University Medical Center Mainz, Mainz, Germany
| | - Philip Wenzel
- Department of Medicine 2 and Center for Thrombosis and Hemostasis University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
46
|
Grieve SM, Mazhar J, Callaghan F, Kok CY, Tandy S, Bhindi R, Figtree GA. Automated quantification of myocardial salvage in a rat model of ischemia-reperfusion injury using 3D high-resolution magnetic resonance imaging (MRI). J Am Heart Assoc 2014; 3:jah3614. [PMID: 25146703 PMCID: PMC4310382 DOI: 10.1161/jaha.114.000956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background Quantification of myocardial “area at risk” (AAR) and myocardial infarction (MI) zone is critical for assessing novel therapies targeting myocardial ischemia–reperfusion (IR) injury. Current “gold‐standard” methods perfuse the heart with Evan's Blue and stain with triphenyl tetrazolium chloride (TTC), requiring manual slicing and analysis. We aimed to develop and validate a high‐resolution 3‐dimensional (3D) magnetic resonance imaging (MRI) method for quantifying MI and AAR. Methods and Results Forty‐eight hours after IR was induced, rats were anesthetized and gadopentetate dimeglumine was administered intravenously. After 10 minutes, the coronary artery was re‐ligated and a solution containing iron oxide microparticles and Evan's Blue was infused (for comparison). Hearts were harvested and transversally sectioned for TTC staining. Ex vivo MR images of slices were acquired on a 9.4‐T magnet. T2* data allowed visualization of AAR, with microparticle‐associated signal loss in perfused regions. T1 data demonstrated gadolinium retention in infarcted zones. Close correlation (r=0.92 to 0.94; P<0.05) of MRI and Evan's Blue/TTC measures for both AAR and MI was observed when the combined techniques were applied to the same heart slice. However, 3D MRI acquisition and analysis of whole heart reduced intra‐observer variability compared to assessment of isolated slices, and allowed automated segmentation and analysis, thus reducing interobserver variation. Anatomical resolution of 81 μm3 was achieved (versus ≈2 mm with manual slicing). Conclusions This novel, yet simple, MRI technique allows precise assessment of infarct and AAR zones. It removes the need for tissue slicing and provides opportunity for 3D digital analysis at high anatomical resolution in a streamlined manner accessible for all laboratories already performing IR experiments.
Collapse
Affiliation(s)
- Stuart M Grieve
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Sydney Translational Imaging Laboratory, Sydney Medical School, University of Sydney, Australia (S.M.G.) Department of Radiology, Royal Prince Alfred Hospital, Sydney, Australia (S.M.G., F.C.)
| | - Jawad Mazhar
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Department of Cardiology, Royal North Shore Hospital, Sydney, Australia (J.M., R.B., G.A.F.)
| | - Fraser Callaghan
- Department of Radiology, Royal Prince Alfred Hospital, Sydney, Australia (S.M.G., F.C.)
| | - Cindy Y Kok
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.)
| | - Sarah Tandy
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.)
| | - Ravinay Bhindi
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Department of Cardiology, Royal North Shore Hospital, Sydney, Australia (J.M., R.B., G.A.F.)
| | - Gemma A Figtree
- North Shore Heart Research Group, Kolling Institute, University of Sydney, Australia (S.M.G., J.M., C.Y.K., S.T., R.B., G.A.F.) Department of Cardiology, Royal North Shore Hospital, Sydney, Australia (J.M., R.B., G.A.F.)
| |
Collapse
|
47
|
Garbers C, Scheller J. Interleukin-6 and interleukin-11: same same but different. Biol Chem 2014; 394:1145-61. [PMID: 23740659 DOI: 10.1515/hsz-2013-0166] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/30/2013] [Indexed: 11/15/2022]
Abstract
The pleiotropic physiological functions of interleukin (IL-)6 type cytokines range from embryonic development and tissue homoeostasis to neuronal development and T cell differentiation. In contrast, imbalance of the well-controlled cytokine signaling network leads to chronic inflammatory diseases and cancer. IL-6 and IL-11 both signal through a homodimer of the ubiquitously expressed β-receptor glycoprotein 130 (gp130). Specificity is gained through an individual IL-6/IL-11 α-receptor, which does not directly participate in signal transduction, although the initial cytokine binding event to the α-receptor leads to the final complex formation with the β-receptors. Both cytokines activate the same downstream signaling pathways, which are predominantly the mitogen-activated protein kinase (MAPK)-cascade and the Janus kinase/signal transducer and activator of transcription (Jak/STAT) pathway. However, recent studies have highlighted divergent roles of the two related cytokines. Here, we will discuss how the biochemical similarities are translated into unique and non-redundant functions of IL-6 and IL-11 in vivo and illustrate strategies for cytokine-specific therapeutic intervention.
Collapse
Affiliation(s)
- Christoph Garbers
- Medical Faculty, Institute of Biochemistry and Molecular Biology II, Heinrich-Heine University, Universit a tsstr. 1, D-40225 Dusseldorf, Germany
| | | |
Collapse
|
48
|
Mesenchymal stem cell therapy for cardiac inflammation: immunomodulatory properties and the influence of toll-like receptors. Mediators Inflamm 2013; 2013:181020. [PMID: 24391353 PMCID: PMC3872440 DOI: 10.1155/2013/181020] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 11/14/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND After myocardial infarction (MI), the inflammatory response is indispensable for initiating reparatory processes. However, the intensity and duration of the inflammation cause additional damage to the already injured myocardium. Treatment with mesenchymal stem cells (MSC) upon MI positively affects cardiac function. This happens likely via a paracrine mechanism. As MSC are potent modulators of the immune system, this could influence this postinfarct immune response. Since MSC express toll-like receptors (TLR), danger signal (DAMP) produced after MI could influence their immunomodulatory properties. SCOPE OF REVIEW Not much is known about the direct immunomodulatory efficiency of MSC when injected in a strong inflammatory environment. This review focuses first on the interactions between MSC and the immune system. Subsequently, an overview is provided of the effects of DAMP-associated TLR activation on MSC and their immunomodulative properties after myocardial infarction. MAJOR CONCLUSIONS MSC can strongly influence most cell types of the immune system. TLR signaling can increase and decrease this immunomodulatory potential, depending on the available ligands. Although reports are inconsistent, TLR3 activation may boost immunomodulation by MSC, while TLR4 activation suppresses it. GENERAL SIGNIFICANCE Elucidating the effects of TLR activation on MSC could identify new preconditioning strategies which might improve their immunomodulative properties.
Collapse
|
49
|
|
50
|
Chemerin15 inhibits neutrophil-mediated vascular inflammation and myocardial ischemia-reperfusion injury through ChemR23. EMBO Rep 2013; 14:999-1007. [PMID: 23999103 PMCID: PMC3818079 DOI: 10.1038/embor.2013.138] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 07/30/2013] [Accepted: 08/02/2013] [Indexed: 02/07/2023] Open
Abstract
Neutrophils are shown to express ChemR23 and respond to C15, which inhibits integrin activation and clustering, reducing neutrophil adhesion and chemotaxis in vitro, and neutrophil recruitment and heart damage in a murine myocardial infarction model. Neutrophil activation and adhesion must be tightly controlled to prevent complications associated with excessive inflammatory responses. The role of the anti-inflammatory peptide chemerin15 (C15) and the receptor ChemR23 in neutrophil physiology is unknown. Here, we report that ChemR23 is expressed in neutrophil granules and rapidly upregulated upon neutrophil activation. C15 inhibits integrin activation and clustering, reducing neutrophil adhesion and chemotaxis in vitro. In the inflamed microvasculature, C15 rapidly modulates neutrophil physiology inducing adherent cell detachment from the inflamed endothelium, while reducing neutrophil recruitment and heart damage in a murine myocardial infarction model. These effects are mediated through ChemR23. We identify the C15/ChemR23 pathway as a new regulator and thus therapeutic target in neutrophil-driven pathologies.
Collapse
|